+ All Categories
Home > Documents > Antithrombotic therapy inatrial fibrillation · 2019-04-12 · Antithrombotic therapy inatrial...

Antithrombotic therapy inatrial fibrillation · 2019-04-12 · Antithrombotic therapy inatrial...

Date post: 25-Jun-2020
Category:
Upload: others
View: 4 times
Download: 0 times
Share this document with a friend
21
Antithrombotic therapy in atrial fibrillation associated with valvular heart disease: a joint consensus document from the European Heart Rhythm Association (EHRA) and European Society of Cardiology Working Group on Thrombosis, endorsed by the ESC Working Group on Valvular Heart Disease, Cardiac Arrhythmia Society of Southern Africa (CASSA), Heart Rhythm Society (HRS), Asia Pacific Heart Rhythm Society (APHRS), South African Heart (SA Heart) Association and Sociedad Latinoamericana de Estimulaci on Card ıaca y Electrofisiolog ıa (SOLEACE) Gregory Y. H. Lip 1 *, Jean Philippe Collet 2 , Raffaele de Caterina 3 , Laurent Fauchier 4 , Deirdre A. Lane 5 , Torben B. Larsen 6 , Francisco Marin 7 , Joao Morais 8 , Calambur Narasimhan 9 , Brian Olshansky 10 , Luc Pierard 11 , Tatjana Potpara 12 , Nizal Sarrafzadegan 13 , Karen Sliwa 14 , Gonzalo Varela 15 , Gemma Vilahur 16 , Thomas Weiss 17 , Giuseppe Boriani 18 and Bianca Rocca 19 ESC Scientific Document Group: Bulent Gorenek 20 (Reviewer Coordinator), Irina Savelieva 21 , Christian Sticherling 22 , Gulmira Kudaiberdieva 23 , Tze-Fan Chao 24 , Francesco Violi 25 , Mohan Nair 26 , Leandro Zimerman 27 , Jonathan Piccini 28 , Robert Storey 29 , Sigrun Halvorsen 30 , Diana Gorog 31 , Andrea Rubboli 32 , Ashley Chin 33 and Robert Scott-Millar 34 * Corresponding author. Tel/fax: þ44 121 5075503. E-mail address: g.y.h.lip@bham.ac.uk Published on behalf of the European Society of Cardiology. All rights reserved. V C The Author 2017. For permissions, please email: [email protected]. Europace (2017) 19, 1757–1758 EHRA CONSENSUS DOCUMENT doi:10.1093/europace/eux240 Downloaded from https://academic.oup.com/europace/article-abstract/19/11/1757/4098134 by guest on 25 August 2018
Transcript
Page 1: Antithrombotic therapy inatrial fibrillation · 2019-04-12 · Antithrombotic therapy inatrial fibrillation associated withvalvular heart disease: a joint consensus document from

Antithrombotic therapy in atrial fibrillation

associated with valvular heart disease: a joint

consensus document from the European

Heart Rhythm Association (EHRA) and

European Society of Cardiology Working

Group on Thrombosis, endorsed by the ESC

Working Group on Valvular Heart Disease,

Cardiac Arrhythmia Society of Southern

Africa (CASSA), Heart Rhythm Society (HRS),

Asia Pacific Heart Rhythm Society (APHRS),

South African Heart (SA Heart) Association

and Sociedad Latinoamericana de

Estimulaci�on Card�ıaca y Electrofisiolog�ıa(SOLEACE)

Gregory Y. H. Lip1*, Jean Philippe Collet2, Raffaele de Caterina3,

Laurent Fauchier4, Deirdre A. Lane5, Torben B. Larsen6, Francisco Marin7,

Joao Morais8, Calambur Narasimhan9, Brian Olshansky10, Luc Pierard11,

Tatjana Potpara12, Nizal Sarrafzadegan13, Karen Sliwa14, Gonzalo Varela15,

Gemma Vilahur16, Thomas Weiss17, Giuseppe Boriani18 and Bianca Rocca19

ESC Scientific Document Group: Bulent Gorenek20 (Reviewer Coordinator),

Irina Savelieva21, Christian Sticherling22, Gulmira Kudaiberdieva23,

Tze-Fan Chao24, Francesco Violi25, Mohan Nair26, Leandro Zimerman27,

Jonathan Piccini28, Robert Storey29, Sigrun Halvorsen30, Diana Gorog31,

Andrea Rubboli32, Ashley Chin33 and Robert Scott-Millar34

* Corresponding author. Tel/fax: þ44 121 5075503. E-mail address: [email protected]

Published on behalf of the European Society of Cardiology. All rights reserved. VC The Author 2017. For permissions, please email: [email protected].

Europace (2017) 19, 1757–1758 EHRA CONSENSUS DOCUMENTdoi:10.1093/europace/eux240

Downloaded from https://academic.oup.com/europace/article-abstract/19/11/1757/4098134by gueston 25 August 2018

Page 2: Antithrombotic therapy inatrial fibrillation · 2019-04-12 · Antithrombotic therapy inatrial fibrillation associated withvalvular heart disease: a joint consensus document from

1Institute of Cardiovascular Sciences, University of Birmingham and Aalborg Thrombosis Research Unit, Department of Clinical Medicine, Aalborg University, Denmark (Chair,representing EHRA); 2Sorbonne Universite Paris 6, ACTION Study Group, Institut De Cardiologie, Groupe Hopital Pitie-Salpetriere (APHP), INSERM UMRS 1166, Paris, France;3Institute of Cardiology, ‘G. D’Annunzio’ University, Chieti, Italy; 4Centre Hospitalier Universitaire Trousseau et Faculte de Medicinde, Universite Francois Rabelais, Tours,France; 5Institute of Cardiovascular Sciences, University of Birmingham, United Kingdom; and Aalborg Thrombosis Research Unit, Department of Clinical Medicine, AalborgUniversity, Aalborg, Denmark; 6Thrombosis Research Unit,Department of Clinical Medicine, Aalborg University Hospital, Aalborg, Denmark; 7Hospital Universitario Virgen de laArrixaca, Murcia, Spain; 8Department of Cardiology, Leiria Hospital Centre, Leiria, Portugal; 9Department of Cardiac Electrophysiology, Care Hospital, Hyderabad, India; 10MercyHospital, Mason City, Iowa, USA; 11Department of Cardiology, University Hospital Sart-Tilman, Liege, Belgium; 12School of Medicine, Belgrade University; Cardiology Clinic,Clinical Center of Serbia, Belgrade, Serbia; 13Isfahan Cardiovascular Research Center (WHO Collaborating Center), Cardiovascular Research Institute, Isfahan University ofMedical Sciences, Isfahan, Iran and School of Population and Public Health, University of British Columbia, Vancouver, Canada; 14Hatter Institute for Cardiovascular Research inAfrica, Faculty of Health Sciences, University of Cape Town, South Africa; and Mary McKillop Institute, ACU, Melbourne, Australia; 15Servicio de Electrofisiolog�ıa, CentroCardiovascular Casa de Galicia, Hidalgos, Uruguay; 16Cardiovascular Science Institute - ICCC, IIB-Sant Pau, CiberCV, Hospital de Sant Pau, Barcelona, Spain; 17MedicalDepartment For Cardiology and Intensive Care, Wilhelminenhospital, and Medical Faculty Sigmund Freud University, Vienna, Austria; 18Cardiology Department, University ofModena and Reggio Emilia, Policlinico di Modena, Modena, Italy; 19Institute of Pharmacology, Catholic University School of Medicine, Rome, Italy (Co-Chair, representing ESCWorking Group on Thrombosis); 20Eskisehir Osmangazi University, Eskisehir, Turkey (Reviewer Coordinator); 21Molecular and Clinical Sciences Institute, St George’s Universityof London, London, UK; 22Department of Cardiology University Hospital Basel, Basel, Switzerland; 23Adana, Turkey; 24Division of Cardiology, Department of Medicine, TaipeiVeterans General Hospital, and Institute of Clinical Medicine, Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan (APHRS reviewer);25University of Rome La Sapienza, Rome, Italy; 26Department of Cardiology, Max Super Specialty Hospital, New Delhi, India; 27Hospital de ClUnicas de Porto Alegre, FederalUniversity of Rio Grande do Sul, Brasil (SOLAECE reviewer); 28Duke University Medical Center, Duke Clinical Research Institute, Durham, USA (HRS reviewer);29Department of Cardiovascular Sciences, University of Sheffield, Sheffield, UK; 31Department of Cardiology, Oslo University Hospital Ulleval, Oslo, Norway; 30National Heartand Lung Institute, Imperial College, London, and Postgraduate Medicine, University of Hertfordshire, Hertfordshire, UK; 32Ospedale Maggiore, Division of Cardiology,Bologna, Italy (Working Group of Thrombosis reviewer); 33Electrophysiology and Pacing, Groote Schuur Hospital, University of Cape Town, South Africa (CASSA reviewer);and 34Department of Medicine, Division of Cardiology, University of Cape Town, South Africa (SAHeart reviewer)

Received 14 June 2017; editorial decision 15 June 2017; accepted 20 June 2017; online publish-ahead-of-print 30 August 2017

Atrial fibrillation (AF) is a major worldwide public health problem, and AF in association with valvular heart disease (VHD) is also com-mon. However, management strategies for this group of patients have been less informed by randomized trials, which have largely focusedon ‘non-valvular AF’ patients.Thrombo-embolic risk also varies according to valve lesion and may also be associated with CHA2DS2VASc score risk factor components,rather than only the valve disease being causal.Given marked heterogeneity in the definition of valvular and non-valvular AF and variable management strategies, including non-vitamin Kantagonist oral anticoagulants (NOACs) in patients with VHD other than prosthetic heart valves or haemodynamically significant mitralvalve disease, there is a need to provide expert recommendations for professionals participating in the care of patients presenting with AFand associated VHD.To address this topic, a Task Force was convened by the European Heart Rhythm Association (EHRA) and European Society ofCardiology (ESC) Working Group on Thrombosis, with representation from the ESC Working Group on Valvular Heart Disease, HeartRhythm Society (HRS), Asia Pacific Heart Rhythm Society (APHRS), South African Heart (SA Heart) Association and SociedadLatinoamericana de Estimulaci�on Card�ıaca y Electrofisiolog�ıa (SOLEACE) with the remit to comprehensively review the published evi-dence, and to publish a joint consensus document on the management of patients with AF and associated VHD, with up-to-date consen-sus recommendations for clinical practice for different forms of VHD.This consensus document proposes that the term ‘valvular AF’ is outdated and given that any definition ultimately relates to theevaluated practical use of oral anticoagulation (OAC) type, we propose a functional Evaluated Heartvalves, Rheumatic or Artificial(EHRA) categorization in relation to the type of OAC use in patients with AF, as follows: (i) EHRA Type 1 VHD, which refers to AF patientswith ‘VHD needing therapy with a Vitamin K antagonist (VKA); and (ii) EHRA Type 2 VHD, which refers to AF patients with ‘VHD need-ing therapy with a VKA or a Non-VKA oral anticoagulant (NOAC)’, also taking into consideration CHA2DS2VASc score risk factor com-ponents.This consensus document also summarizes current developments in the field, and provides general recommendations for the managementof these patients based on the principles of evidence-based medicine....................................................................................................................................................................................................

Keywords Atrial fibrillation • Valvular heart disease • Mitral stenosis • Mechanical prosthetic heart valves • Thrombo-embolism • Stroke • Stroke prevention • Anticoagulation • Vitamin K antagonists • Non-vitamin K antago-nist oral anticoagulants • Pregnancy

1758 G.Y.H. Lip

Downloaded from https://academic.oup.com/europace/article-abstract/19/11/1757/4098134by gueston 25 August 2018

Page 3: Antithrombotic therapy inatrial fibrillation · 2019-04-12 · Antithrombotic therapy inatrial fibrillation associated withvalvular heart disease: a joint consensus document from

Preamble and valvular heartdisease definition

Atrial fibrillation (AF) is a major public health problem1 with globalprevalence rates (per 1000000 population) in 2010 being 596.2(95% uncertainty interval (UI), 558.4–636.7) in men and 373.1(95% UI, 347.9–402.2) in women; the incidence rates increasedto 77.5 (95% UI, 65.2–95.4) in men and 59.5 (95% UI, 49.9–74.9)in women.2 Worldwide, AF in association with valvular heart dis-ease (VHD) is also common, and management strategies forthis group of patients have been less addressed by randomizedtrials. The latter have largely focused on ‘non-valvular AF’ patientsleading to major uncertainties over how to define (and treat) suchpatients.

There is also an important heterogeneity in the definition of valvu-lar and non-valvular AF.3 Some physicians assume that any valve dis-ease should be considered as ‘valvular’ AF. Others consider that onlymechanical valve prosthesis and rheumatic mitral stenosis should bedefined as ‘valvular’ AF.

The term valvular AF has been arbitrarily applied and the 2016ESC guidelines have avoided the term ‘valvular AF’ and refer sim-ply to ‘AF related to hemodynamically significant mitral stenosisor prosthetic mechanical heart valves’.4 AF clearly leads to anincremental risk for thromboembolism in patients with mitralvalve stenosis, but there are limited data for other valvular dis-eases. Another proposal is to use the acronym MARM-AF as asimple acronym to designate ‘Mechanical and Rheumatic MitralAF’ as an alternative to term ‘valvular AF’ to designate the clinicalscenarios for which at the non-vitamin K antagonist oral anticoa-gulants (NOACs) are not indicated.5

For this document we recognize the uncertainty in terminology,and our scope largely relates to AF related to ‘hemodynamically sig-nificant’ rheumatic VHD (ie. severe enough to impact on patient’ssurvival or necessitates an intervention or surgery) or prostheticmechanical heart valves. Nonetheless, thrombo-embolic (TE) riskvaries according to valve lesion and may be associated withCHA2DS2VASc score risk factor components, rather than the valvedisease per se being causal.6,7 TE risk may also be influenced not onlyby type but also the severity of the lesion. For example, the degree ofmitral regurgitation may matter when it comes to risk of TE as somestudies suggest that mild (Grade 1) mitral regurgitation is associatedwith a 2.7-fold increased risk of stroke/TE, while severe forms maypossibly have a ‘protective’ effect (HR = 0.45 for stroke and 0.27 forLA stasis.8 An appropriate definition of ‘valvular AF’ would need toidentify a subgroup of patients with similar pathophysiology ofthrombo-embolism, TE risk, and treatment strategies6,9; however,this would be challenging given the major heterogeneity of thecondition.

This consensus document proposes that the term ‘valvular AF’ isoutdated and given that any definition ultimately relates to the eval-uated practical use of oral anticoagulation (OAC) type, we propose afunctional EHRA (Evaluated Heartvalves, Rheumatic or Artificial) cat-egorization in relation to the type of OAC use in patients with AF, asfollows:

Evaluated Heartvalves, Rheumatic or Artificial (EHRA) Type 1,which refers to AF patients with ‘VHD needing therapy with aVitamin K antagonist (VKA)’

• Mitral stenosis (moderate-severe, of rheumatic origin)• Mechanical prosthetic valve replacement

[EHRA Type I VHD is broadly similar to the previously describedMARM-AF.]

Evaluated Heartvalves, Rheumatic or Artificial (EHRA) Type 2VHD, which refers to AF patients with ‘VHD needing therapy with aVKA or a NOAC’, also taking into consideration CHA2DS2VAScscore risk factor components:

• Mitral regurgitation• Mitral valve repair• Aortic stenosis• Aortic regurgitation• Tricuspid regurgitation• Tricuspid stenosis• Pulmonary regurgitation• Pulmonic stenosis• Bioprosthetic valve replacements• Trans-aortic valve intervention (TAVI)

This classification would have the advantage that it may easilyevolve or be updated (type 1 may become type 2 or vice versa)when there are new results. For example, transcatheter mitral valveinterventions (TMVI, e.g. to include both MitraClip and Mitral valvereplacement) are emerging as a possible therapeutic options,10 butmore data are awaited especially in relation to OAC use.

A recent physicians survey3 reported marked heterogeneity in thedefinition of valvular and non-valvular AF and variable managementstrategies, including NOACs in patients with VHD other than pros-thetic heart valves or haemodynamically significant mitral stenosis.Thus, there is a need to provide expert recommendations for profes-sionals participating in the care of patients presenting with AF andassociated VHD. These may include rheumatic VHD, mechanical orbiological prosthetic valves and percutaneous aortic valve implanta-tion (TAVI), as well as those having undergone mitral valve repair.Whilst hypertrophic cardiomyopathy is sometimes discussed in asso-ciation with valvular AF, this would not be addressed in this document,given specific guidelines on the management of this condition.11

To address this topic, a Task Force was convened by the EuropeanHeart Rhythm Association (EHRA) and European Society ofCardiology (ESC) Working Group on Thrombosis, with representa-tion from the ESC Working Group on Valvular Heart Disease, HeartRhythm Society (HRS), Asia Pacific Heart Rhythm Society (APHRS),South African Heart (SA Heart) Association and SociedadLatinoamericana de Estimulaci�on Card�ıaca y Electrofisiolog�ıa(SOLEACE) with the remit to comprehensively review the publishedevidence, and to publish a joint consensus document on the manage-ment of patients with AF and associated VHD, with up-to-date con-sensus recommendations for clinical practice.

This document summarizes current developments in the field, andprovides general recommendations for the management of thesepatients based on the principles of evidence-based medicine.

Antithrombotic therapy in AF with valvular heart disease 1758a

Downloaded from https://academic.oup.com/europace/article-abstract/19/11/1757/4098134by gueston 25 August 2018

Page 4: Antithrombotic therapy inatrial fibrillation · 2019-04-12 · Antithrombotic therapy inatrial fibrillation associated withvalvular heart disease: a joint consensus document from

Rather than ‘valvular AF’ a more functional Evaluated Heartvalves,Rheumatic or Artificial (EHRA) categorization in relation to the typeof OAC use is recommended in patients with AF, as follows:

(i) EHRA Type 1 VHD, which refers to AF patients with ’VHDneeding therapy with a Vitamin K antagonist (VKA); and

(ii) EHRA Type 2 VHD, which refers to AF patients with‘VHD needing therapy with a VKA or a Non-VKA oralanticoagulant (NOAC), also taking into considerationCHA2DS2VASc score risk factor components.

Evidence reviewMembers of the Task Force were asked to perform a detailed litera-ture review, weigh the strength of evidence for or against a particulartreatment (or procedure), and include estimates of expected healthoutcomes where data exist. Patient-specific modifiers, comorbidities,and issues of patient preference that might influence the choice ofparticular tests or therapies are considered, as are frequency offollow-up and cost effectiveness. In controversial areas, or withregard to issues without evidence other than usual clinical practice, aconsensus was achieved by agreement of the expert panel after thor-ough deliberations. This document was prepared by the Task Forcewith representation from EHRA, HRS, APHRS, and SOLAECE. Thedocument was peer-reviewed by official external reviewers repre-senting EHRA, HRS, APHRS, and SOLAECE.

Consensus statements are evidence-based, and derived primarilyfrom published data. Current systems of ranking level of evidence arebecoming complicated in a way that their practical utility might be com-promised.12 We have, therefore, opted for an easier and, perhaps, moreuser-friendly system of ranking that should allow physicians to easilyassess current status of evidence and consequent guidance (Table 1).

Thus, a ‘green heart’ indicates a recommended statement or rec-ommended/indicated treatment (or procedure) and is based on atleast one randomized trial, or is supported by large observational evi-dence that it is beneficial and effective. A ‘yellow heart’ indicates gen-eral agreement and/or scientific evidence favouring a statement orthe usefulness/efficacy of a treatment or procedure. A yellow heartmay be supported by randomized trials based on small number ofpatients or not widely applicable. Treatment strategies for whichthere have been scientific evidence that they are potentially harmfuland should not be used are indicated by a ‘red heart’.

EHRA grading of consensus statements does not have separatedefinitions of Level of Evidence. The categorisation used for consen-sus statements (used in consensus documents) should not be consid-ered as being directly similar to that used for official society guidelinerecommendations which apply a classification (I–III) and level of evi-dence (A, B, and C) to recommendations in official guidelines.

Finally, this is a consensus document that includes evidence andexpert opinions from several countries. The anticoagulationapproaches discussed may, therefore, include drugs that do not havethe approval of governmental regulatory agencies in all countries.

Relationships with industry and otherconflictsIt is EHRA/ESC policy to sponsor position papers and guidelineswithout commercial support, and all members volunteered their

time. Thus, all members of the writing group as well as reviewershave disclosed any potential conflict of interest in detail, at the end ofthis document.

Epidemiology of valvular atrialfibrillation and implications forstroke/thrombo-embolism

The reported prevalence of AF varies by geographical region.2 InAustralian, North American and European studies, approximately1–2% of adults have AF.13–15 In Asians, the reported prevalence ofAF ranges from 0.1–4.0% in the community and 2.8–14% in hospital-based studies.16

Nonetheless, robust data on the epidemiology of patients with AFand associated VHD are limited. Examples of available data fromsome global registries and large trials are reported in Supplementarymaterial online, Table S1. In the RE-LY AF Registry which enrolledpatients presenting to an emergency department with AF at 164 sitesin 46 countries, rheumatic heart disease was present in 2.2% of NorthAmerican patients, in comparison with 21.5% in Africa and 31.5% inIndia7; interestingly thrombo-embolism rates were related to clinicalrisk profile, as expressed by CHADS2 score, irrespective of the pres-ence of rheumatic VHD. Detailed data on the geographic distributionof valvular AF are also reported in the Supplementary material online.

.................................................................................................

Table 1 Scientific rationale of recommendations

Definitions where related

to a treatment or

procedure

Consensus

statement

Symbol

Scientific evidence that a treat-

ment or procedure is benefi-

cial and effective. Requires at

least one randomized trial, or

is supported by strong obser-

vational evidence and authors’

consensus (as indicated by an

asterisk).

Recommended/

indicated

General agreement and/or scien-

tific evidence favour the use-

fulness/efficacy of a treatment

or procedure. May be sup-

ported by randomized trials

based on small number of

patients or not widely

applicable.

May be used or

recommended

Scientific evidence or general

agreement not to use or rec-

ommend a treatment or

procedure.

Should NOT be

used or

recommended

This categorization for our consensus document should not be considered asbeing directly similar to that used for official society guideline recommendationswhich apply a classification (I–III) and level of evidence (A, B, and C) torecommendations.

1758b G.Y.H. Lip

Downloaded from https://academic.oup.com/europace/article-abstract/19/11/1757/4098134by gueston 25 August 2018

Page 5: Antithrombotic therapy inatrial fibrillation · 2019-04-12 · Antithrombotic therapy inatrial fibrillation associated withvalvular heart disease: a joint consensus document from

Pathophysiology—a brief overview

There is a general agreement that AF is independently associated withthrombotic diathesis.17 The drivers include the three elements of theVirchow’s triad: blood flow alterations, endocardial injury and changes inblood constituents.18 In fact, according to the recently published EHRA/HRS/APHRS/SOLAECE consensus document, atrial tissue in VHD ischaracterized, at a histopathological level, by a combination of cardio-myocyte and fibrotic changes.19 Co-morbidities present in most AFpatients may contribute to enhance thrombotic risk by pro-inflamma-tory/pro-atherogenic mechanisms. An overview of the pathophysiology

of thrombogenesis in AF in haemodynamically significant mitral stenosisand/or mechanical heart valves prosthesisis shown in Figure 1.

The risk of TE is increased in patients with AF and mechanicalvalve, mild-to-severe mitral stenosis20 and left atrium dilatation, ascompared to non-valvular AF,6 suggesting differences among thepathogenic mechanisms contributing to thrombosis in each of theseAF conditions. It is generally thought that Virchow’s triad is triggeredby the turbulent flow and the endothelial injury that accompaniesvalvular AF. On top of this, AF prosthetic valves (particularly mechan-ical prosthesis) induce thrombin generation through the activation ofboth the tissue factor (TF) and the contact coagulation pathways.21

Figure 1 Pathophysiology of thrombogenesis in atrial fibrillation (AF) related prosthesis and/or mitral valve diseases. In valvular-AF there is a propen-sity to thrombosis because of the presence of the Virchow’s triad components which, in turn, are found likely boosted by patients’ co-morbid conditions.The risk of thrombosis, however, is enhanced because of the presence of prosthetic valves which activate the coagulation cascade (both the intrinsic andextrinsic pathway) leading to thrombin production (a strong platelet agonist); and, although to a lesser extent, because of the considerable degree ofmitral stenosis which induces flow turbulences capable of inducing platelet activation. Finally, AF also frequently occurs in patients with severe aorticstenosis, which can be associated to the Heyde’s syndrome due to Von Willebrand Factor (VWF) consumption leading to an acquired bleeding disorder.

Antithrombotic therapy in AF with valvular heart disease 1758c

Downloaded from https://academic.oup.com/europace/article-abstract/19/11/1757/4098134by gueston 25 August 2018

Page 6: Antithrombotic therapy inatrial fibrillation · 2019-04-12 · Antithrombotic therapy inatrial fibrillation associated withvalvular heart disease: a joint consensus document from

Surgical heart valve replacement surgery induces tissue damage withTF release leading to extrinsic coagulation pathway activation afterbinding to plasma Factor (F) VII/FVIIa. Moreover, the exposure ofvalve leaflets, struts and/or sewing ring to the circulating blood22 canactivate the contact (intrinsic) coagulation pathway. Both intrinsicand extrinsic pathways converge factor X (FX) activation and thenthe transformation of prothrombin into thrombin (FIIa) and forma-tion of the fibrin mesh. The vitamin K antagonist (VKA), warfarin, byblocking the formation of the vitamin K-dependent clotting FVII, FIX,FX, and FII prevents the activation of the coagulation cascade at theextrinsic and intrinsic pathway levels.

In addition to the thrombogenic contribution of plasma coagula-tion in valvular AF, platelet activation may possibly contribute,although to a much lesser extent, to promote thrombogenesis, par-ticularly in mild-to-severe mitral stenosis. It is also possible that mod-erate to severe mitral stenosis causes increased blood stasis, due tothe severe dilatation of the left atrium occurring in this condition.Finally, acquired type IIA von Willebrand disease and bleeding com-plications can be associated with severe aortic stenosis due to high-molecular weight multimer consumption.23

Oral anticoagulation with vitaminK antagonists in patients with AFand prosthetic heart valves,including bioprostheses

Mechanical heart valvesOral anticoagulation (OAC) with VKA is crucial for prevention of TEin patients with mechanical heart valves, regardless the presence orabsence of AF. The ESC guidelines24 establish the risk of TE in patientswith mechanical valves according to valve type and position, and alsoaccording to the individual patient risk profile or comorbidities.Warfarin and other VKA are the most widely used OACs, and aretitrated according to international normalized ratio (INR) range andtarget value which is also related with associated risk factors (Table 2).

The duration of antithrombotic therapy also varies according to anumber of factors. Lifelong anticoagulant treatment is indicated for allpatients with mechanical valves and those with bioprosthetic valvesor native valve disease with >_1 additional risk factors.

Clinical trials with VKA are sparse and recommendations aremostly based on old trials, with the oldest types of mechanical valveprostheses, and AF patients were excluded.

Patients with bioprostheses and additional risk factors for embo-lism (AF, venous TE, hypercoagulable state, or with a lesser degree ofevidence, severely impaired left ventricular function) require life-longOAC. The use of NOACs instead of warfarin in this setting isaccepted by the more recent document of recommendations byEHRA25 in spite of a lack of randomized clinical trials (RCT)26–28 (seeAntithrombotic therapy in patients with AF undergoing TAVI orLAAO section for NOACs and bioprosthesis).

BioprosthesesAfter biological valve replacement, thrombo-embolic risk is esti-mated between 0.6 to 3.3% per year without anticoagulation, after

the third month.29 The thrombo-embolic risk associated with a bio-prosthesis and sinus rhythm is higher in the first 3 months after sur-gery, the risk being almost eliminated in anticoagulated patients foraortic bioprosthesis, but remaining higher in patients with a mitralbioprosthesis.30,31 The benefit of an initial anticoagulant treatmentfollowing aortic valve replacement with a bioprosthesis and no AF ishowever debated.32–34

Overall AF patients with a bioprosthesis had a non-significantlyhigher risk of stroke/TE events compared with patients with non-valvular AF, and VKA use was independently associated with alower risk of thrombo-embolic events (hazard ratio 0.83, 95%CI 0.71–0.98).35

One small pilot study compared Dabigatran Versus Warfarin AfterBioprosthesis Valve Replacement for the Management of AtrialFibrillation Postoperatively (DAWA Pilot Study) but small numberspreclude definitive conclusions.36

Recent small studies also suggest that NOACs can be a reasonablealternative to VKA in patients with AF and remote bioprostheticvalve implantation,26,37 however larger studies are needed to definethe safety and efficacy profile. Data on thromboprophylaxis inpatients with AF and TAVI, which is actually the insertion of a bio-prosthesis, are preliminary38 and discussed in detail elsewhere in thedocument section.

Mitral valve repairPatients undergoing mitral valve repair have a small risk of TE,39 withthe highest risk of TE occurring during the first year after surgery.Guidelines therefore recommend OAC during the first 3–6 monthsafter surgery.40 However, only limited data are available on the effi-cacy of warfarin therapy in the early stages after valve surgery, andthe use of short-term VKAs in patients with mitral valve annuloplastyis also controversial. It is therefore not clear whether patients withAF in addition to valve repair are markedly different from the patientswith AF and no VHD.6,21

North American and European guidelines have a different positionon this issue, the former considering AF is ‘valvular AF’ whilst ESCguidelines do not.32,41

.................................................................................................

Table 2 Target international normalized ratio forsome examples of mechanical prosthesis

Prosthesis

thrombogenecity

Valve type Patient-related

risk factors*

Risk factor >_ 1

Low Carbomedics, Medtronic

Hall, St Jude Medical,

ON-X

3.0

Medium Other bileaflet valves 3.5

High Lillehei-Kaster,

Omniscience, Starr-

Edwards, Bjork-Shiley

and other tilting-disc

valves

4.0

*Risk factors: previous thromboembolism; AF; mitral stenosis of any degree; leftventricular ejection fraction < 35%. Reproduced from reference.24

1758d G.Y.H. Lip

Downloaded from https://academic.oup.com/europace/article-abstract/19/11/1757/4098134by gueston 25 August 2018

Page 7: Antithrombotic therapy inatrial fibrillation · 2019-04-12 · Antithrombotic therapy inatrial fibrillation associated withvalvular heart disease: a joint consensus document from

Consensus statements

Indications of ‘add on’ antiplatelettherapy in patients with atrialfibrillation and prostheticmechanical heart valves

Arterial TE and valve thrombosis are �12%/year and 22%/year, formechanical valve prosthesis in the aortic and mitral position, respec-tively, in patients without VKA prophylaxis.29 The residual risk rangesfrom 0.5%/year, to 2.5%/year,29,42–44 in VKA-treated patients withoutadditional cardiovascular risk factors such as AF. A higher incidence isassociated with the mitral (�2%/year) vs. the aortic (�1%/year) posi-tion, depending also on the type of valve and VKA intensity.29,43,44 AFand/or other risk factors (e.g. heart failure, even without AF) increaseTE risk by four-fold, from 4% up to 8%/year,45–47 even on adequateVKA treatment.45,46

Given this high residual TE risk, RCT have compared VKA alone vs.VKA combined with different aspirin doses and/or dipyridamole29,48,49

(Table 3).Despite major methodological limitations of these studies includ-

ing small sample size, heterogeneities in thrombotic risk level at studyentry and anticoagulation intensity, inconsistencies in safety and effi-cacy endpoint definitions,48 there may possibly be some benefit ofadding low-dose aspirin (between 75 and 200 mg daily) to VKA inpatients with mechanical valve prosthesis and additional risk factorsincluding AF29,48,54 (Table 3). This approach lowered TE complica-tions in the majority of studies,29,45,46,48,54 and two meta-analysesshowed�60% relative risk reduction (RRR) of TE and�50% RRR ofall-cause mortality48,49 (Table 3). Nonetheless, the relative risk ofmajor bleeding with VKAs plus antiplatelet therapy increases by�58% across studies including aspirin daily doses from 100 to1000 mg48,49 and high dose dipyridamole alone or with aspirin.48

Importantly, major bleeding appears significantly affected by aspirindose: the association with low dose (100 mg) shows a bleeding risksignificantly lower than higher doses50,52 and not significantly differentfrom VKA alone (OR = 0.96; 95% CI 0.60–1.55; 2.58; 95% CI 1.43–2.55 for low and high doses vs. VKA, respectively, P = 0.002 for thehigh aspirin dose combination vs. VKA) with similar efficacy(Table 3).48,50 Thus, VKA plus low-dose aspirin (75–100 mg daily) forthe association of mechanical prosthetic valve and AF, is

recommended by the AHA/ASA/ACCP as a class I (level A or B) rec-ommendation,42,44,55,56 but as a class IIb C recommendation by theESC.

When the aspirin/VKA combination is used, anticoagulation shouldbe titrated taking into account the type of valve, the position, andcomorbidities. The target INR for AF patients with aortic mechanicalprosthetic valve on VKA and low-dose aspirin should be 2.5 (range2.0–3.0), with close attention to the quality of anticoagulation con-trol, with time in therapeutic range (TTR) >65–70%. This applies tolow-thrombogenicity valves, including Carbomedics, Medtronic Hall,St Jude Medical, ON-X; but there is insufficient data for other valves,even if bileaflet.

Whether the INR target should be 2.5 (range 2–3) or 3 (range2.5–3.5) in AF patients with mitral prosthetic valve on both VKA andlow-dose aspirin is less clear. High intensity VKA (i.e. INR range 3–4or higher), combined with aspirin, has been consistently associatedwith higher major bleeding and comparable benefit as lower intensityVKA with aspirin.51,52,57

Consensus statements

Evidence for non-vitamin Kantagonist oral anticoagulants usein patients with atrial fibrillationand valvular heart disease

Subgroups from the recent non-vitaminK antagonist oral anticoagulant trialsThe efficacy and safety of NOACs for the prevention of stroke/sys-temic embolic events (SSEE) in patients with non-valvular AF hasbeen established by the pivotal randomized trials.58,60–63 These trialsexcluded patients with significant mitral stenosis or prostheticmechanical valves but enrolled participants (13–26%, depending on

.................................................................................................

Coloured

heart

Supporting

references

• Well managed VKA monotherapy with

good anticoagulation control (e.g. TTR

>65–70%), is generally recommended,

taking into account the type of valve, the

position, and additional risk factor(s),

including atrial fibrillation.

24

• Patients with a bioprosthetic valve and

atrial fibrillation require lifelong OAC.

.................................................................................................

Coloured

heart

Supporting

references

• In patients with a mechanical pros-

thetic valve and concomitant AF with

vascular disease, VKA plus low-dose

aspirin (75–100 mg daily) may be con-

sidered in the absence of high bleed-

ing risk.

53,54,56–69

� In patients with a mechanical pros-

thetic valve and AF, when VKA plus

aspirin are used, the INR should be

kept between 2.0 and 3.0 (target 2.5),

given the high bleeding risk of the

combination and the lack of evidence

of greater protection with higher

intensity VKA (INR range 3–5 or

above).

51,56,63

• High doses of aspirin (>_325 mg) in

association with VKA at any intensity

must be avoided.

51,60

Antithrombotic therapy in AF with valvular heart disease 1758e

Downloaded from https://academic.oup.com/europace/article-abstract/19/11/1757/4098134by gueston 25 August 2018

Page 8: Antithrombotic therapy inatrial fibrillation · 2019-04-12 · Antithrombotic therapy inatrial fibrillation associated withvalvular heart disease: a joint consensus document from

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

.

Tab

le3

Ran

do

miz

ed

tria

lsan

dm

eta

-an

aly

ses

co

mp

ari

ng

dif

fere

nt

inte

nsi

ties

ofV

KA

alo

ne

vs.

VK

Ap

lus

asp

irin

inp

ati

en

tsw

ith

mech

an

icalvalv

ere

pla

cem

en

tsi

nce

1990

Refe

ren

ce

Desi

gn

Pati

en

tsA

Fan

dM

VR

po

siti

on

(%)

Eff

icacy

Safe

tyC

om

men

tso

rad

di-

tio

nald

ata

Tur

pie

etal

.45

RC

T:V

KA

(INR

targ

et

3–4.

5)þ

ASA

100

mg

daily

vs.V

KA

(INR

targ

et

3–4.

5)þ

plac

ebo

mea

nf.u

.2.5

year

s

370

patie

nts

with

MV

Ror

tissu

eva

lve

repl

acem

ent

þA

For

TE;

mea

nag

e58

year

s

mal

es50

%

AF:

45%

per

arm

;

Mitr

al44

%,

Aor

tic46

%

Mul

tiple

10%

Maj

orsy

stem

icem

bolis

m

orva

scul

arde

ath:

1.9%

/

year

ASA

vs.8

.5%

/yea

r

plac

ebo

NN

15

Maj

orbl

eedi

ng:8

.5%

/yea

r

ASA

,6.6

%/y

ear

plac

ebo

P=

0.43

;

NN

52

Mea

nIN

R:3

inea

char

m.

Net

bene

ficia

leffe

ctfa

v-

oure

dA

SA(R

RR

61%

)

Altm

an

etal

.50

RC

T:V

KA

(INR

targ

et2–

3)

plus

:ASA

100

mg

orA

SA

650

mg

daily

mea

nf.u

.20

mon

ths

416

patie

nts

with

mec

hani

-

calM

VR

;

mea

nag

e60

year

s

mal

es50

%

AF:�

23%

per

arm

Mitr

al26

%,

Aor

tic74

%

Maj

orT

E:0.

5%/y

ear

inlo

w-

ASA

and

1.1%

/yea

rhi

gh

ASA

;

Vas

cula

rde

ath

1.2%

/yea

r

low

-ASA

vs.0

.5%

/yea

r

high

-ASA

,P=

0.3

Maj

orbl

eedi

ng:3

.6%

/yea

r

low

-ASA

vs.5

.1%

/yea

r

high

-ASA

;

any

blee

ding

:7.9

%/y

ear

low

-ASA

vs.1

3.4%

/yea

r

high

-ASA

,

p=

0.03

;

Vas

cula

rm

orta

lity

and

non-

fata

lTE:

3.4%

low

-

ASA

and

2.9%

high

ASA

,

P=

n.s.

Mes

chen

gies

-

eret

al.5

1

RC

T:h

igh

inte

nsity

VK

A

(INR

3.5-

4.5)

vs.l

ess

inte

nse

VK

A(IN

R2.

5-3.

5)

þA

SA10

0m

g

med

ian

f.u.:

23m

onth

s

503

patie

nts

with

MR

V;

�30

%m

itral

MV

R

med

ian

age

53ye

ars

mal

es58

%

AF:

15%

high

VK

Aar

m;2

0%

VK

ASA

arm

;p=

0.04

Mitr

al29

%

Aor

tic66

%

Mul

tiple

4%

Maj

orT

E:1.

48%

/yea

rhi

gh

VK

Avs

.1.3

2%/y

ear

VK

A

þA

SA,

P=

n.s.

Maj

orbl

eedi

ng:2

.23%

/yea

r

high

VK

Avs

.1.1

3%/y

ear

VK

ASA

;p=

ns

GIb

leed

ing:

2.12

%/y

ear

high

VK

Avs

.0.7

6%/y

ear

VK

ASA

,

P=

0.14

Mea

nIN

R:h

igh

VK

Aar

m

3.98

.

VK

ASA

3.1

p<

0.00

1

Mor

talit

y:hi

ghV

KA

8.1%

vs.

VK

ASA

3.5%

(RR

for

VK

ASA

0.41

,95%

CI

0.23

-0.8

1)62

Laffo

rtet

al.4

6R

CT

:VK

ASA

200

mg

daily

vs.V

KA

alon

e

INR

targ

et3

(2.5

-3.5

)f.u

.:

1ye

ar

229

patie

nts

with

MV

R;

mea

nag

e63

year

s

mal

es50

%

Non

-sin

usrh

ythm

:

�49

%pe

rar

m

Mitr

al:1

00%

Maj

orT

E:0.

9%A

SAþ

VK

A

vs.4

.1%

VK

A,P

=ns

.

Tot

alT

E:9%

ASAþ

VK

A

vs.2

5%V

KA

,P=

0.00

4;

NN

T¼6

Echo

card

iogr

aphi

cth

rom

bi

day

9:4.

8%A

SAþ

VK

Avs

.

13.1

%V

KA

,P=

0.03

Maj

orbl

eedi

ng:1

9.2%

ASAþ

VK

Avs

.8.3

%V

KA

,

P=

0.02

NN

10

No

sign

ifica

ntdi

ffere

nces

inm

orta

lity

(few

even

ts)

Lars

onan

d

Fish

er49

Met

a-an

alys

isof

4tr

ials

usin

gas

piri

n

869

patie

nts

Var

iabl

ede

pend

ing

onth

e

stud

y

Maj

orT

E:3.

5%V

KAþ

ASA

vs.1

1.3%

VK

Aon

ly

NN

T:13

Maj

orbl

eedi

ng:1

3.1%

VK

A

þA

SAvs

.8.1

%V

KA

only

NN

H:2

0

All-

caus

em

orta

lity:

5.4%

VK

ASA

vs.7

.9%

VK

A

only

Asp

irin

daily

dose

rang

ed

from

100

to10

00m

g

Cont

inue

d

1758f G.Y.H. Lip

Downloaded from https://academic.oup.com/europace/article-abstract/19/11/1757/4098134by gueston 25 August 2018

Page 9: Antithrombotic therapy inatrial fibrillation · 2019-04-12 · Antithrombotic therapy inatrial fibrillation associated withvalvular heart disease: a joint consensus document from

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

.

Tab

le3

Co

nti

nu

ed

Refe

ren

ce

Desi

gn

Pati

en

tsA

Fan

dM

VR

po

siti

on

(%)

Eff

icacy

Safe

tyC

om

men

tso

rad

di-

tio

nald

ata

Peng

oet

al.5

2R

CT

:low

-inte

nsity

VK

A

(INR

2–3)þ

ASA

100

mg

vs.h

ighe

rin

tens

ityV

KA

(INR

2.5–

3.5)

for

6m

onth

s

f.u.:

1.5

year

s

198

patie

nts

with

MV

R;

mea

nag

e60

year

s

mal

es46

%

�28

%pe

rar

m

Mitr

al28

%

Aor

tic63

%

Mul

tiple�

10%

VK

ASA

:4m

ajor

blee

d-

ing

and

1is

chae

mic

stro

ke;V

KA

:2m

ajor

blee

ding

and

2is

chae

mic

stro

ke,P

=0.

6

Cum

ulat

ive

endp

oint

of

maj

orbl

eedi

ngan

d

thro

mbo

sis

Ver

ysm

alls

tudy

with

shor

t

trea

tmen

tan

dlo

wnu

m-

ber

ofev

ents

Don

get

al.5

3R

CT

:VK

ASA

75–

100

mg

vs.V

KA

alon

e

mea

nf.u

.:24

±9

mon

ths

1496

patie

nts

with

mec

hani

calM

VR

;

mea

nag

e35

year

s

mal

es40

%

AF:

40%

per

arm

Mitr

al83

%

Aor

tic43

%

Mul

tiple

:16%

Maj

orT

E:2.

1%V

KAþ

ASA

vs.3

.6%

VK

Aal

one,

P=

0.04

NN

66

Maj

orbl

eedi

ng:3

.5V

KAþ

ASA

vs.3

.7%

inV

KA

alon

e;P

=ns

NN

500

War

fari

ndo

se:2

.92

±

0.87

mg

inV

KAþ

ASA

and

2.89

±0.

79m

gin

VK

A

alon

e.

No

diffe

renc

esin

mor

talit

y

rate

Mas

sela

nd

Litt

le48

Met

a-an

alys

isof

RC

Tco

m-

pari

ngV

KA

alon

evs

.

VK

Aan

dan

tipla

tele

ts

4122

patie

nts

with

MV

Rin

aort

icor

mitr

alpo

sitio

n

orbo

th

Var

iabl

ede

pend

ing

onth

e

stud

y

Maj

orT

E:A

SAþ

VK

Avs

.

VK

Aal

one

OR

0.43

[95%

CI0

.32-

0.5]

P<

0.00

1

Maj

orbl

eedi

ng:A

ntip

late

let

þV

KA

vs.V

KA

alon

eO

R

1.58

[95%

CI1

.14–

2.18

]

P<

0.00

1

ASA

high

:OR

2.58

[95%

CI

1.43

–1.5

5]

ASA

100

mg:

OR

0.96

[95%

CI0

.6–1

.55]

Stat

istic

alin

tera

ctio

nhi

gh

vs.l

owP

=0.

04

Ove

rall

mor

talit

y:

OR

0.57

[95%

CI0

.42-

0.78

]

Maj

orbl

eedi

ngin

stud

ies

pre-

1990

:OR

2.34

[95%

CI1

.34–

4.08

]

afte

r-19

90:O

R1.

26[9

5%

CI0

.84–

1.89

]

Dat

aar

epr

esen

ted

as%

/yea

r,w

hene

ver

poss

ible

.NN

Tan

dN

NH

per

year

are

calc

ulat

edfo

rth

eco

mpa

riso

nsin

clud

ing

the

com

bine

dV

KAþ

antip

late

let

trea

tmen

tvs

.VK

Aal

one,

whe

neve

rpo

ssib

le.

AF,

atri

alfib

rilla

tion;

ASA

,asp

irin

;f.u

.,fo

llow

-up;

INR

,int

erna

tiona

lnor

mal

ized

ratio

;MV

R,m

echa

nica

lval

vere

plac

emen

t;N

NT

,num

ber

need

edto

trea

t;N

NH

,num

ber

need

edto

harm

;OR

,odd

sra

tio;R

CT

,ran

dom

ized

clin

ical

tria

l;R

RR

,re

lativ

eri

skre

duct

ion;

TE,

thro

mbo

embo

lism

;VK

A,v

itam

inK

anta

goni

sts.

Antithrombotic therapy in AF with valvular heart disease 1758g

Downloaded from https://academic.oup.com/europace/article-abstract/19/11/1757/4098134by gueston 25 August 2018

Page 10: Antithrombotic therapy inatrial fibrillation · 2019-04-12 · Antithrombotic therapy inatrial fibrillation associated withvalvular heart disease: a joint consensus document from

the trial)28,59,64,65 with other clinically significant non-rheumatic VHD,including mitral regurgitation (MR), aortic regurgitation (AR), aorticstenosis (AS), mild mitral stenosis (MS) or prior valve surgery (bio-prosthetic valves or valve repair) (Table 4). There are limited or nodata on other options, such as MitraClip or other TMVI, and thus,NOACs should not be used in these patients.

Variable inclusion/exclusion criteria across the NOACs trialsreflect the prevailing lack of a clear-cut definition of valvular AF.6

Patients with VHD of non-rheumatic origin are prevalent in clinicalpractice,66 and physicians may often deny NOACs to eligible AFpatients due to uncertainty over whether the patient has valvular ornon-valvular AF.3

There are no randomized trials on NOACs in AF patients withVHD. In the Randomized Evaluation of Long-Term AnticoagulationTherapy (RE-LY),59 Rivaroxaban Once Daily Oral Direct Factor XaInhibition Compared with Vitamin K Antagonism for Prevention ofStroke and Embolism Trial in Atrial Fibrillation (ROCKET-AF),64

Apixaban for Reduction in Stroke and Other ThromboembolicEvents in Atrial Fibrillation (ARISTOTLE)65 and EffectiveAnticoagulation with factor Xa Next Generation in Atrial Fibrillation-Thrombolysis in Myocardial Infarction 48 (ENGAGE AF-TIMI 48)28

trial subgroup analyses, non-valvular AF patients with VHD wereolder, had more comorbidities (including renal dysfunction), morepersistent/permanent AF and higher cardioembolic and bleeding risksthan patients without VHD. Whilst the use of aspirin was broadly

similar, prior VKA use was more common among patients with VHD.Irrespective of the treatment arm (i.e. warfarin or a NOAC), VHDpatients generally experienced worse outcomes (stroke and systemicTE, major bleeding or all-cause death) in comparison to non-VHDpatients (Table 5). Nonetheless, the efficacy of NOACs in reductionof SSEE or all-cause mortality was consistent among patients withoutor with VHD (irrespective of the VHD subtype). Likewise, the safetyof NOACs in terms of lower risk of major bleeding or ICH was con-sistent irrespective of VHD status, excluding the significantly higherrates of major bleeding in VHD patients (particularly those withaortic stenosis, or mitral or aortic regurgitation)69 treated with rivar-oxaban compared to warfarin (Table 5). Importantly, there are nohead-to-head comparisons for any NOAC vs. VKA in AF patientswith moderate to severe mitral stenosis; as mentioned, these patientswere not enrolled in the NOACs trials.

The number of patients with any prior valve surgery (i.e. bio-prosthetic valves or valve repair) exposed to rivaroxaban, apixa-ban or edoxaban in the ROCKET-AF, ARISTOTLE or ENGAGEAF-TIMI 48 trials, respectively, was very low (Table 4).Nevertheless, as reported for apixaban and edoxaban,26,68 therewas no statistically significant interaction between the presence ofa bioprosthetic heart valve and the respective NOAC effects(Table 5), thus suggesting that apixaban or edoxaban may possiblybe alternatives to warfarin in AF patients with bioprosthetic valvesimplanted >_3 months ago.

....................................................................................................................................................................................................................

Table 4 Inclusion/exclusion criteria pertinent to valvular heart disease in the pivotal NOAC trials in patients with‘non-valvular’ AF and valvular disease type distribution across the trials

Inclusion (�)/exclusion (–) criteria RE-LY61 ROCKET-AF58 ARISTOTLE60 ENGAGE-AF62 AVERROES63

Prosthetic heart valve(s)

Mechanical – – – – –

Bioprosthesis – – � � �Prior surgical repaira – � � � �Moderate-to-severe MS – – – – –

Other significant valve diseaseb – � � � –

Mild-to-moderate valve disease � � � � �

Subgroups with a cardiac valve diseasec RE-LY58 ROCKET-AF64 ARISTOTLE67 ENGAGE-AF28

Total n (%) 3950 (21.8) 2003 (14.1) 4808 (26.4) 2824 (13.4) NR

Moderate/severe MR 3101 (78.5) 1756 (87.7) 3526 (73.3) 2250 (79.6) NR

Moderate/severe AR 817 (20.7) 486 (24.3) 887 (18.4) 369 (13.0) NR

Moderate/severe AS 471 (11.9) 215 (10.7) 384 (8.0) 165 (5.8) NR

Other 1179 (6.5) 11 (0.6)d 2124 (44.2) NR NR

Mild MS 193 (4.9) NR 131 (2.7) 254 (9.0) NR

Prior valve surgery (excluding mechanic

prosthetic heart valve)

Not applicable 106 (5.3) 251 (5.2) 325 (11.5) NR

Valve repair — 42 (2.1%) NR 123 (4.3) NR

Valvuloplasty — 64 (3.2%) NR 19 (0.7) NR

Bioprosthetic valves — Not applicable 82 (1.7) 191 (6.8) NR

AF, atrial fibrillation; AR, aortic regurgitation; AS, aortic stenosis; MR, mitral regurgitation; MS, mitral stenosis; NOAC, non-vitamin K antagonist oral anticoagulant; NR, notreported.aAnnuloplasty, Commisurotomy, Valvuloplasty, etc.bClinically significant, but not requiring immediate surgery repair.cCategories are not mutually exclusive.dWithout any of the preceding.

1758h G.Y.H. Lip

Downloaded from https://academic.oup.com/europace/article-abstract/19/11/1757/4098134by gueston 25 August 2018

Page 11: Antithrombotic therapy inatrial fibrillation · 2019-04-12 · Antithrombotic therapy inatrial fibrillation associated withvalvular heart disease: a joint consensus document from

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

....

Tab

le5

Majo

ro

utc

om

es

inA

Fp

ati

en

tsb

yvalv

ula

rh

eart

dis

ease

statu

san

dtr

eatm

en

t

Ou

tco

me

tria

lN

OA

C/W

arf

ari

nV

HD

(Rate

%/y

ear)

No

VH

D

(Rate

%/y

ear)

VH

DN

OA

Cvs.

warf

ari

nH

R(9

5%

CI)

No

VH

DN

OA

Cvs.

warf

ari

nH

R(9

5%

CI)

Inte

racti

on

P

Stro

ke/S

E

RO

CK

ET-A

F64

Riv

arox

aban

/War

fari

n2.

01/2

.43

1.96

/2.2

20.

83(0

.55–

1.27

)0.

89(0

.75–

1.07

)0.

70

AR

IST

OT

LE65

Api

xaba

n/W

arfa

rin

1.46

/2.0

81.

20/1

.43

0.70

(0.5

1–0.

97)

0.84

(0.6

7–1.

04)

0.37

8

RE-

LY59

Dab

i-150

mg/

War

fari

n1.

12/1

.90

1.11

/1.6

60.

59(0

.37–

0.93

)0.

67(0

.52–

0.86

)0.

63

Dab

i-110

mg/

War

fari

n1.

84/1

.90

1.45

/1.6

60.

97(0

.65–

1.45

)0.

88(0

.70–

1.10

)0.

65

ENG

AG

E-A

F28

HD

ER/W

arfa

rin

1.39

/2.0

2

1.94

/2.0

2

1.60

/1.7

7

2.04

/1.7

7

0.69

(0.4

4–1.

07)

0.97

(0.6

6–1.

44)

0.91

(0.7

7–1.

07)

1.15

(0.9

8–1.

35)

0.25

5

0.44

0LD

ER/W

arfa

rin

AR

IST

OT

LE67

biop

rost

hetic

valv

es(n

=82

)A

pixa

ban/

War

fari

n2.

9/0.

0N

AN

RN

A—

ENG

AG

E-A

F26

biop

rost

hetic

valv

es(n

=19

1)H

DER

/War

fari

n1.

19/1

.66

NA

0.37

(0.1

0–1.

42)

NA

0.15

LDER

/War

fari

n2.

57/1

.66

NA

0.53

(0.1

6–1.

78)

NA

0.31

Maj

orbl

eedi

ng

RO

CK

ET-A

F64

Riv

arox

aban

/War

fari

n6.

14/4

.20

3.22

/3.3

31.

56(1

.14–

2.14

)0.

98(0

.84–

1.15

)0.

01

AR

IST

OT

LE65

Api

xaba

n/W

arfa

rin

2.49

/3.1

42.

01/3

.07

0.79

(0.6

1–1.

04)

0.65

(0.5

5–0.

77)

0.23

RE-

LY59

Dab

i-150

mg/

War

fari

n4.

21/5

.12

3.06

/3.1

40.

82(0

.64–

1.06

)0.

98(0

.83–

1.15

)0.

25

Dab

i-110

mg/

War

fari

n3.

77/5

.12

2.63

/3.1

40.

73(0

.56–

0.95

)0.

84(0

.71–

0.99

)0.

38

ENG

AG

E-A

F28

HD

ER/W

arfa

rin

3.28

/4.4

6

1.82

/4.4

6

2.66

/3.2

7

1.59

/3.2

7

0.74

(0.5

3–1.

02)

0.41

(0.2

8–0.

60)

0.82

(0.7

1–0.

94)

0.49

(0.4

1–0.

57)

0.57

3

0.43

9LD

ER/W

arfa

rin

AR

IST

OT

LEbi

opro

sthe

ticva

lves

(n=

82)6

8A

pixa

ban/

War

fari

n7.

9/5.

2N

AN

RN

A0.

61

ENG

AG

E-A

F29

biop

rost

hetic

valv

es(n

=19

1)H

DER

/War

fari

nN

RN

A0.

50(0

.15–

1.67

)N

A0.

26

LDER

/War

fari

n0.

76/6

.27

NA

0.12

(0.0

1–0.

95)

NA

0.04

5

ICH

b

RO

CK

ET-A

F64

Riv

arox

aban

/War

fari

n0.

88/0

.73

0.43

/0.7

41.

27(0

.58–

2.79

)0.

59(0

.40–

0.86

)0.

084

AR

IST

OT

LE65

Api

xaba

n/W

arfa

rin

0.25

/0.8

80.

37/0

.78

0.28

(0.1

4–0.

57)

0.47

(0.3

3–0.

68)

0.20

RE-

LY63

Dab

i-150

mg/

War

fari

n0.

34–0

.93

0.31

/0.7

20.

36(0

.17–

0.77

)0.

43(0

.28–

0.67

)0.

68

Dab

i-110

mg/

War

fari

n0.

27/0

.93

0.21

/0.7

20.

29(0

.13–

0.68

)0.

30(0

.18–

0.49

)0.

98

ENG

AG

E-A

F28

HD

ER/W

arfa

rin

0.32

/0.8

2

0.24

/0.8

2

0.41

/0.8

5

0.26

/0.8

5

0.39

(0.1

5–0.

98)

0.29

(0.1

1–0.

79)

0.48

(0.3

5–0.

66)

0.31

(0.2

1–0.

45)

0.65

7

0.92

6LD

ER/W

arfa

rin

All-

caus

em

orta

lity

RO

CK

ET-A

F64

Riv

arox

aban

/War

fari

n5.

48/5

.60

4.19

/4.6

00.

98(0

.75–

1.29

)0.

91(0

.80–

1.03

)0.

60

AR

IST

OT

LE65

Api

xaba

n/W

arfa

rin

4.95

/4.8

83.

02/3

.61

1.01

(0.8

4–1.

22)

0.84

(0.7

3–0.

96)

0.10

1

RE-

LY59

Dab

i-150

mg/

War

fari

n4.

28/4

.73

3.46

/3.9

60.

91(0

.70–

1.17

)0.

87(0

.75–

1.01

)0.

79

Dab

i-110

mg/

War

fari

n4.

35/4

.73

3.58

/3.9

60.

92(0

.71–

1.19

)0.

90(0

.78–

1.04

)0.

89

ENG

AG

E-A

F28

HD

ER/W

arfa

rin

LDER

/War

fari

n6.

46/5

.71

5.81

/5.7

1

3.62

/4.1

3

3.46

/4.1

3

1.13

(0.9

0–1.

41)

1.03

(0.8

2–1.

29)

0.88

(0.7

8–0.

98)

0.83

(0.7

5–0.

93)

0.04

5

0.10

0

AR

IST

OT

LEbi

opro

sthe

ticva

lves

(n=

82)6

9A

pixa

ban/

War

fari

n6.

9/7.

1N

AN

RN

A0.

88

ENG

AG

E-A

F26 b

biop

rost

hetic

valv

es(n

=19

1)H

DER

/War

fari

nN

RN

A0.

46(0

.23–

0.91

)N

A0.

03

LDER

/War

fari

nN

RN

A0.

43(0

.21–

0.88

)N

A0.

02

ICH

,int

racr

ania

lhae

mor

rhag

e;N

A,n

otap

plic

able

;NO

AC

S,no

n-vi

tam

inK

oral

antic

oagu

lant

s;N

R,n

otre

port

ed;S

E,sy

stem

icem

bolis

m;V

HD

,val

vula

rhe

art

dise

ase.

a The

rew

asno

effe

ctm

odifi

catio

nof

the

pres

ence

orab

senc

eof

VH

Don

rela

tive

outc

omes

with

high

er-

orlo

wer

-dos

eed

oxab

anin

com

pari

son

tow

arfa

rin

(all

inte

ract

ion

pw

ere

non-

sign

ifica

nt).

bIn

the

suba

naly

ses

incl

udin

gon

lybi

opro

sthe

ticva

lves

,the

rate

sof

ICH

wer

eno

tsp

ecifi

ed;C

ompo

site

outc

ome

ofst

roke

/SE,

maj

orbl

eedi

ngor

deat

h.

Antithrombotic therapy in AF with valvular heart disease 1758i

Downloaded from https://academic.oup.com/europace/article-abstract/19/11/1757/4098134by gueston 25 August 2018

Page 12: Antithrombotic therapy inatrial fibrillation · 2019-04-12 · Antithrombotic therapy inatrial fibrillation associated withvalvular heart disease: a joint consensus document from

A meta-analysis70 of the VHD sub-groups from the RE-LY,ROCKET-AF, ARISTOTLE and ENGAGE AF-TIMI 48 trials broadlyconfirmed the findings shown in Table 5. Overall, AF patients withVHD had non-significantly higher rate of SSEE (RR 1.13; 95% CI,0.99–1.28) and significantly higher rates of major bleeding (RR 1.34;95% CI, 1.13–1.49) and all-cause death (RR 1.34; 95% CI, 1.13–1.59)than patients without VHD.

Compared with warfarin, the use of NOACs (i.e. rivaroxaban,apixaban or higher doses of dabigatran or edoxaban) was associatedwith consistently lower rates of SSEE regardless of the presence orabsence of VHD (RR 0.70; 95% CI, 0.58–0.86 and 0.84; 95% CI, 0.75–0.95, respectively; interaction P = 0.31), similar major bleeding rates(VHD RR 0.93; 95% CI, 0.67–1.27 and no-VHD RR 0.85; 95%CI 0.70–1.02, interaction P = 0.63), consistently lower rates of ICH(VHD RR 0.47; 95% CI, 0.24–0.93 and no-VHD RR 0.49; 95%CI, 0.41–0.59, interaction P = 0.91) and higher all-cause death rate inVHD patients (RR 1.01; 95% CI, 0.90–1.14) than in those withoutVHD (RR 0.88; 95% CI, 0.82–0.94), interaction P = 0.03,70 In the anal-ysis that also included the lower doses of dabigatran and edoxaban,the magnitude of SSEE risk reduction with NOACs vs. warfarin wasslightly reduced, as well as the rates of major bleeding and ICH, butthere were no significant subgroup interactions by VHD status.Overall, the presence of VHD did not affect the relative protectiveeffect of NOACs compared with warfarin in terms of SSEE andmajor bleeding. These findings were further supported by anothermeta-analysis of the four NOACs yielding identical results.71 Of note,both meta-analyses reported significant treatment effect heterogene-ity regarding the analysis of major bleeding.

With the exclusion of patients with moderate-to-severe mitralstenosis, prosthetic mechanical heart valve, TAVI or TMVI, whowere not enrolled in the non-valvular AF trials, the aforemen-tioned subgroup and meta-analyses may suggest that AF patientswith VHD would experience at least the same benefit fromNOACs as patients without VHD. However, due to limitationsinherent to these types of analyses, further RCTs are required inAF patients with VHD before recommendations can be given (seeTables 4 and 5).

Prosthetic mechanical heart valves:Randomized, Phase II Study to Evaluatethe Safety and Pharmacokinetics of OralDabigatran Etexilate in Patients afterHeart Valve ReplacementMechanical valve prosthesis trigger complex mechanisms of throm-bogenesis and are associated with a very high cardioembolic riskrequiring chronic OAC even in the absence of AF. Animal studies onmechanical valve implantation using first the direct FIIa inhibitors mel-agatran72 and then dabigatran73,74 as well as the phase III data fromthe RE-LY trial61 informed the only study to date on a NOAC inpatients with mechanical heart valves.

The Randomized, Phase II Study to Evaluate the Safety andPharmacokinetics of Oral Dabigatran Etexilate in Patients afterHeart Valve Replacement (RE-ALIGN) trial was a phase-II, con-trolled, dose-finding, open-label study75 randomizing (2:1)patients with aortic (n = 172; 68%) or mitral (n = 71; 28%) mechan-ical valve replacement, or both (n = 9; 4%) to dabigatran or

adjusted-dose warfarin (target INR 2.0–3.0 or 2.5–3.5 in aortic ormitral position, respectively). The initial dabigatran dose of 150,220, or 300 mg b.i.d. (selected according to renal function) wasfurther adjusted over 12 weeks to achieve the primary study out-come—a trough plasma concentration >_50 ng/mL, based on thepharmacokinetic model from the RE-LY trial. Most patients (79%)received study drug 5–7 days after surgery, and 23% of patientshad AF. The RE-ALIGN study was prematurely terminated afterrandomizing 252 of 405 planned patients, due to an excess instroke (5% vs. 0%), valve thrombosis (3% vs. 0%) and major bleed-ing events (4% vs. 2%) in the dabigatran arm, after a mean dabiga-tran exposure of �20 weeks. Different explanations have beenproposed, including inadequate dabigatran plasma concentrations,different pharmacodynamics of dabigatran and warfarin, excessiveactivation of the contact coagulation pathway induced by the sew-ing ring in the early postoperative course, a higher inter-individualvariability in the dabigatran arm and differences in predicted vs.observed drug concentrations in the RE-LY vs. RE-ALIGN.75 Arecent in vitro study suggested that the dabigatran trough plasmaconcentration required to reduce valve-induced FIIa generationshould be much higher than 50 ng/mL (that is, 260 ng/mL) corre-sponding to a 620 mg b.i.d. dosing.68 At present, all AF patientswith a mechanical valve prosthesis should be treated with VKAsConsensus statements

.................................................................................................

Coloured

heart

Supporting

references

• The use of the NOACs in patients

with AF and mechanical valve pros-

thesis is contraindicated.

76

• Randomized clinical trials testing the

efficacy and safety of direct oral FXa

inhibitors in patients with AF and

mechanical heart valved prosthesis

are lacking. Until more data are avail-

able, rivaroxaban, apixaban, and edox-

aban are contraindicated in such

patients.

. . .

• Until more data are available, AF

patients with any degree of rheumatic

mitral valve stenosis and those with

moderate-to-severe non-rheumatic

mitral stenosis should not be treated

with NOACs.

. . .

• The efficacy and safety of NOACs for

stroke/SE prevention may be similar

in AF patients with and without con-

servative valve surgery such as annu-

loplasty, commissurotomy or

valvuloplasty, or bioprosthetic valves

based on small numbers of patients in

post hoc analyses of RCTs. More data

9,66,72,77,78

Continued

1758j G.Y.H. Lip

Downloaded from https://academic.oup.com/europace/article-abstract/19/11/1757/4098134by gueston 25 August 2018

Page 13: Antithrombotic therapy inatrial fibrillation · 2019-04-12 · Antithrombotic therapy inatrial fibrillation associated withvalvular heart disease: a joint consensus document from

Antithrombotic therapy inpatients with atrial fibrillationundergoing trans-aortic valveintervention or left atrialappendage occlusion

Trans-aortic valve interventionprocedureMost ischaemic events after TAVI are cerebrovascular, and for theseAF is a strong contributor.77 AF is common among high-risk patientswith severe aortic stenosis undergoing TAVI, and is associated with a>2-fold increased risk of all-cause and cardiovascular death, irrespec-tive of the type of AF.78 In addition, the implanted valve adds aprothrombotic environment, which would accentuate the cardioem-bolic risk. Of importance, the gradient of risk directly correlates withthe CHA2DS2-VASc score, which is usually used to aid decision-making as whether to initiate OAC.77 At least 30% of the TAVI popu-lation requires OAC,1 a strategy that seems underused and that hasnever been evaluated prospectively. Dual antiplatelet therapy(DAPT) remains the most widely used antithrombotic strategy afterTAVI, being used in >60% of patients, while VKA is used in <20% ofpatients,77 although AF is observed in >40% of TAVI patients.Current recommendations are expert-driven, rather than evidence-based (Table 6).

Up to 35% patients undergo coronary stenting prior to TAVI. Insuch patients, the risk of stent thrombosis and/or ischaemic cardiacevents in addition to that of AF should be considered in the overallrisk assessment.82 Here triple therapy, a combination of a VKA, low-dose aspirin and clopidogrel, is used in high risk patients, and associ-ated with an increased risk of death, stroke, TE, or major bleedingwhen compared to VKA alone.78,82 Such combinations should be

discussed in the context of recent (i.e. <6 months) acute coronarysyndrome and/or stent implantation, especially in the presence of anunfavourable coronary anatomy (more than three stents, stentlength >_60 mm, multivessel disease, left main disease) but should beavoided whenever deemed possible given the established bettersafety and the possible preserved efficacy of a combination of war-farin and clopidogrel in patients with AF undergoing drug-elutingstent placement.83–85

Recent evidence suggests that VKA alone is much safer and pro-vides a similar rate of ischaemic events as compared to VKA plus anti-platelet therapy (aspirin) in patients undergoing TAVI.82 Howeverthis study was observational, not randomized with an unbalancednumber of patients per treatment arm, and randomized confirmationis needed. Therefore, the association of OAC with SAPT in AFpatients who underwent successful TAVI should be considered up toone year when there is a recent ACS or a recent coronary stenting86

and when the bleeding risk is deemed low (Figure 2).OAC alone as antithrombotic strategy is currently being tested

in three trials (POPular-TAVI NCT02247128, GALILEONCT02556203, ATLANTIS trial NCT02664649), while another trialis testing aspirin alone or in combination with clopidogrel (ARTENCT02640794), although AF patients appear excluded. Indeed, thebenefit of VKA over DAPT in AF depends on the quality of INR con-trol,76 and it has been modelled that a time in therapeuticrange >_58% would be needed to benefit from being on OAC ratherthan on DAPT,76 which is probably not the case in the TAVIpopulation.

Subclinical valve thrombosis is another challenging issue as it maymay occur early after TAVI. Although the frequency of this poten-tially ominous phenomenon remains undefined, as this condition isdifficult to detect, but it seems reversible with anticoagulation.Whether it is associated with cerebrovascular events remains to beestablished.87 Given all these uncertainties, ongoing trials are alsotesting the anticoagulation hypothesis after successful TAVI irrespec-tive of the need of OAC hypotheses using NOACs (NCT02556203,NCT02664649) which have been shown to be better tolerated.Figure 2 shows all currently recommended treatment options.

Recent observational evidence suggest the safety of FXa inhibitionin TAVI,88 showing the feasibility of NOAC in the post-TAVI setting.However, results from randomized comparison of FXa inhibition vs.other antithrombotic strategies are lacking.

Left atrial appendage occlusionprocedureFollowing clinical trials,89–91 percutaneous endovascular left atrialappendage occlusion (LAAO) has been increasingly developed andperformed worldwide for patients with AF, especially those withcontraindications to long-term OAC.92,93 This is supported by guide-lines from the ESC, which give a class IIB recommendation for LAAOin AF patients with high stroke risk and contraindications to long-term OAC.4

Antithrombotic therapy following LAAO has not been well eval-uated, and it is not even known whether OAC or antiplatelet therapyor no therapy is preferable. When possible according to the patientbleeding risk profile, after LAAO most centres use a 6-week periodof VKA (target INR 2.5) followed by once daily clopidogrel (75 mg)

.................................................................................................

Continued

Coloured

heart

Supporting

references

are needed to define the role of

NOACs in this setting.• The efficacy and safety of NOACs in

patients with non-rheumatic mitral

and/or aortic regurgitation or other

native VHD may be similar to AF

patients without VHD based on small

numbers of patients in post hoc analy-

ses of RCTs. More data are needed to

define the role of NOACs in this

setting.

9,66–72,77,78

• In patients with haemodynamically

insignificant valve disease and in those

who have had prior successful balloon

mitral valvulotomy, NOACs can be

considered as substitute for VKAs.

67,68,71–75,77

NB. Recommendations for TAVI are given in the specific section.

Antithrombotic therapy in AF with valvular heart disease 1758k

Downloaded from https://academic.oup.com/europace/article-abstract/19/11/1757/4098134by gueston 25 August 2018

Page 14: Antithrombotic therapy inatrial fibrillation · 2019-04-12 · Antithrombotic therapy inatrial fibrillation associated withvalvular heart disease: a joint consensus document from

and aspirin (75–325 mg) until the 6-months visit. Some patients mayalso receive NOAC.94 Subsequently, low-dose aspirin alone iscontinued indefinitely, as tested in the pivotal trials.89,90,95 This anti-platelet regimen has never been compared with any long termNOAC-based anticoagulation regimen.9,96 However, the ASAP studyshowed that LAAO with the Watchman device is feasible and could

be safely performed without OAC cover (but with antiplatelet ther-apy).95 Such strategy is being evaluated in the ongoing ADRIATICstudy (Apixaban versus Dual or single antiplatelet therapy to ReduceIschemic and bleeding events in Atrial fibrillation patients Treatedwith Invasive Closure of the left atrial appendage). The ASAP TOOrandomized trial (NCT02928497) is currently establishing the safety

....................................................................................................................................................................................................................

Table 6 Recommendations for antithrombotic therapy during and after TAVI in the guidelines in patients with andwithout indication for OAC

ACC/AHA/STS79 ESC80 ACCP44 CCS44,80

Procedural Unfractionated Heparin

(ACT> 300 s)

– –

Post-procedural Aspirin 81 mg indefinitely

and Clopidogrel 75 mg

for 3 up to 6 months

Aspirin (or clopidogrel)

indefinitely

Aspirin (50–100 mg/day)

and Clopidogrel

(75 mg/day) in the first

3 months

Low-dose Aspirin indefi-

nitely and 1–3 months

of a thienopyridine

(no evidence).

Aspirin and clopidogrel

in first 3 months after

TAVI

Patients with a clear

indication for OAC (as

in AF)

It is reasonable to con-

tinue low-dose Aspirin,

but other antiplatelet

therapy should be

avoided

No antiplatelet therapy

but OAC alone

No recommendation Adjunctive antiplatelet

agents is controversial

and triple therapy

should be avoided

ACC, American College of Cardiology; ACT, activated clotting time; ACCP, American College of Chest Physicians; AF, atrial fibrillation; AHA, American Heart Association;ASA, acetylsalicylic acid (aspirin); AVR, aortic valve replacement; CCS, Canadian Cardiovascular Society; ESC, European Society of Cardiology; INR, international normalizedratio; VKA, vitamin K antagonists.

Figure 2 Proposed algorithm for AF patients undergoing a TAVI procedure (adapted from references 87). O refers to oral anticoagulation as VKA(or possibly NOAC). ASC, acute coronary syndrome; CAD, coronary artery disease; DAPT, dual antiplatelet therapy; OAC, oral anticoagulant;SAPT, single antiplatelet therapy.

1758l G.Y.H. Lip

Downloaded from https://academic.oup.com/europace/article-abstract/19/11/1757/4098134by gueston 25 August 2018

Page 15: Antithrombotic therapy inatrial fibrillation · 2019-04-12 · Antithrombotic therapy inatrial fibrillation associated withvalvular heart disease: a joint consensus document from

and effectiveness of the LAAO vs. SAPT in patients with non-valvularAF deemed not to be eligible for OAC to reduce the risk of stroke.Consensus statements

Antithrombotic therapy forvalvular atrial fibrillation inpregnant women

Valvular AF in pregnancy is relatively rare and can be due to congenitalheart disease, mitral valve prolapse with significant mitral regurgitation,or to rheumatic heart disease. Valves can be repaired or replaced witha mechanical valve prosthesis.97 Pregnancy by itself is a prothromboticstate and the coalescence of venous stasis and hypercoagulabilityresults in nearly a five-fold increase in the risk of venous thromboemb-olism during pregnancy.86 The goal of anticoagulation during pregnancyshould be to safely balance the maternal risk of TE and haemorrhagewith the foetal risk of exposure to VKA. The continuously changingpharmacokinetics of LMWH during the various trimesters adds anadditional challenge and requires monitoring by peak and trough anti-Xa levels,86 which is often not feasible (Figure 3).

Women of child-bearing age with VHD need to be comprehen-sively counselled prior to valve replacement and pre-pregnancy todecide on the most appropriate type of valve and to be made awareof the teratogenicity and fetotoxicity of VKA, pregnancy-induced hae-modynamic changes and the pre-existing hypercoagulable statewhich can compromise foetal development and significantly increasethe risk of serious and/or fatal complications to both mother andchild.98 Women with mechanical prosthetic valves should ideally

have preconception evaluation, including advice on risk predictionand contraception, by a joint cardiac-obstetric team seeking advicefrom other specialties.97 Careful counselling on maternal and off-spring risk should be done according the modified World HealthOrganization classification and should include information on compli-cations such as heart failure, valve thrombosis, bleeding complicationswhich can occur during, but also beyond the immediate deliveryperiod. Also, the consequences of the medication that may berequired (for example warfarin embryopathy) need to be discussed.However, often women in some countries may present after20 weeks of gestation, which has implications for their functionalassessment, harmful medication can’t be terminated timeously andlimits the option for pregnancy termination. Such cases are challeng-ing and should be managed at tertiary centre where they can beappropriately assessed and guided/treated. Since anticoagulation isrecommended in pregnant women with AF at risk of stroke, to mini-mise teratogenic risk and intrauterine bleeding, the ESC guidelinesrecommend that dose adjusted heparin should be used during thefirst trimester of pregnancy and in the 2–4 weeks before delivery.4

VKA or heparin can be used in the remaining trimesters of the preg-nancy.4 In the absence of adequate safety data, NOACs should beavoided in pregnancy and in women planning a pregnancy4.Consensus statements

Patient values and preferences,and societal issues

Treatment decisions need to balance the benefits and risks of treat-ment and manage realistic patient expectations, particularly in associ-ation with co-morbidities and in pregnancy. These decisions arecomplex and require assimilation of life expectancy, ability and will-ingness to take anticoagulants, risk of bleeding, lifestyle, co-morbidities, risk of re-operation, and patient preference.100–102

Clinical guidelines on the management of VHD100 advocate incor-porating informed patient preferences into treatment decisions andtechnological advances (for VHD) must be employed ‘responsiblywithin a framework of care which enables shared decision makingand promotes patient goals and well-being’.103 This requires candiddiscussions between the patient and physician to ensure that

.................................................................................................

Coloured

heart

Supporting

references

• AF patients who underwent successful

TAVI may be treated with FXa inhibitors;

however data are limited.

9

• AF patients with stable coronary artery

disease who underwent TAVI may be

treated with OAC only, including VKA

and FXa inhibitors; however prospective

data are limited.

9,82,83

• Based on trial protocols, OAC and single

antiplatelet therapy after successful

LAAO may be used up to 6 weeks in low

bleeding risk patients, followed by single

antiplatelet therapy; however, long term

data are limited, nor any comparison with

NOACs.

89,90,95

• Single antiplatelet therapy or no antith-

rombotic therapy may be used after

LAAO in AF patients who are not eligible

for VKA; however, long term data are lim-

ited, nor any comparison with NOACs.

9,96

.................................................................................................

Coloured

heart

Supporting

references

• There is no consensus on the optimal

regimen for anticoagulation in peri-

partum women with mechanical valve

prosthesis with AF.

86,97,99

• As the optimal anticoagulation regi-

men for use in pregnancy and peripar-

tum remains undetermined, all

decisions should be made by a fully

informed mother and partner in con-

sultation with a multidisciplinary team.

86,97,99

Antithrombotic therapy in AF with valvular heart disease 1758m

Downloaded from https://academic.oup.com/europace/article-abstract/19/11/1757/4098134by gueston 25 August 2018

Page 16: Antithrombotic therapy inatrial fibrillation · 2019-04-12 · Antithrombotic therapy inatrial fibrillation associated withvalvular heart disease: a joint consensus document from

treatment is not futile. Shared decision making103,104 requires patientsto be appropriately informed about treatment options and likely out-comes, to have the type of patient-physician relationship wherepatients feel able to ask questions and where physicians provideinformation and communicate risk effectively,105,106 to enablepatients to make an informed decision incorporating their values,goals and preferences.103,107,108 Patient’s treatment preferences arelikely to vary markedly, with patients often willing to accept higherlevels of risk.108 Patient decision-aids including micro-simulationmodels are available for patients with VHD.109,110

Implications for low-to-middle incomecountriesValvular AF is more common in the Asian and African populationcompared to their western counterparts mainly due to greater bur-den of rheumatic heart disease.110,111,112 Stroke risk is higher amongpatients with valvular AF (17–18%/year) compared to those withnon-valvular AF (4%/year).113 Further, AF may further increase therisk of bioprosthetic valve thrombosis (see Supplementary materialonline, Table S4).114 The burden of rheumatic valve disease is higher,but the quality of anticoagulation is suboptimal in low and middleincome countries. Monitoring of the INR and follow-up remains poorand significant proportion of patients present with sub-therapeutic

INR. The majority of these patients are young (median age28 years),118 unemployed (75.3%) and women (51–66%)112,115 ofreproductive age. On average, they tend to be nearly 10–12 yearsyounger than their western counterparts. Many are unaware of theconcept of therapeutic range INR (60%) and few (<4%) are on con-traceptives despite treatment with warfarin. The NOACs are expen-sive and beyond the reach of the majority of patients requiring themin these countries. Suboptimal anticoagulation and consequentincreased risk of stroke, may lead to significant disability adjusted lifeyears lost and this is likely to pose a major economic burden.Strategies to improve awareness: (i) about the disease, (ii) medicationside-effects, (iii) importance of medication adherence and INR moni-toring, and 4) the danger of anticoagulation during pregnancy arescanty. Although point of care INR testing shows promise (seeSupplementary material online, Table S4), its use among patients fromthe developing world needs to be determined. The impact ofNOACs is less certain, although one recent Brazilian study evaluatingNOACs in public health system context found that NOACs presenta lower cumulative cost per patient when compared to VKAs.116

Health economic perspectivesAF is a disease that induces significant consumption of resources andcosts, encompassing direct medical costs, associated with patient’s

Figure 3 Management strategies for women with mechanical valve prosthesis. CCF, congestive heart failure; ICU, Intensive Care Unit.

1758n G.Y.H. Lip

Downloaded from https://academic.oup.com/europace/article-abstract/19/11/1757/4098134by gueston 25 August 2018

Page 17: Antithrombotic therapy inatrial fibrillation · 2019-04-12 · Antithrombotic therapy inatrial fibrillation associated withvalvular heart disease: a joint consensus document from

medical care (hospitalisations, medications, out-patient visits, etc.),and direct non-medical costs (i.e. costs related to residential or socialcare, as well as out of pocket expenses).117,118

Other costs that are usually taken into account in health-economic analyses are productivity losses caused by patients’ inabilityto work, or absence from work of relatives in order to provide infor-mal care.119 In patients with AF direct costs, reported as per-patientannual costs, have been estimated $2000 to 14 200 in NorthAmerica and 450 to 3000 Euros in Europe per patient.119

Patients with VHD who have AF require appropriate risk stratifica-tion for stroke/SE and, when indicated, the consequent prescriptionof OAC implies a difficult balance between the risk of stroke and sys-temic TE and the risk of bleeding.120,121 Stroke and major bleedinghave also an economic effect. Indeed, the main drivers of costs in AFpatients are AF-related hospitalizations, stroke and haemorrhagicevents. For strokes occurring in patients with AF, the direct costs perpatient are approximately 33% greater than the costs of stroke not-related to AF126 and are in the range of 30 000 Euros over a 2-yearperiod for a severe ischaemic stroke.123 The costs of intra-cerebralhaemorrhage is 50% higher than the cost of ischaemic stroke over a1 year time course.124

Underutilization of, and non-adherence to, warfarin is also quitecommon and is associated with increased costs,125,126 resulting fromTE and haemorrhagic complications. Improved adherence to OAC inAF patients at risk of stroke is important in order to attain the fullclinical and economic benefit of thromboprophylaxis.

Non-vitamin K antagonist oral anticoagulants can be prescribed tosome subgroups of patients with VHD6,127,128 and a series of analysesfocusing on the cost-effectiveness of these agents vs. warfarin hasbeen published, although no study considered separately patientswith VHD. In general, despite the higher cost of NOACs as com-pared to warfarin, the associated benefits make these agentscost-effective in the long-term, especially in settings with poor antico-agulation control associated with VKAs.129,130

Consensus statements

Summary and areas for futureresearch

Mechanical valve prosthesesCurrently, patients with AF and a mechanical prosthesis should onlybe treated with a VKA. Since the RE-ALIGN study, no other NOAC(factor Xa inhibitor drug class) has been tested in this patient group.75

However, the thrombotic risk could be reduced once endothelial tis-sue is present around the ring.17 A trial could potentially be designedafter endothelialization: the first 3 months with VKA, followed by arandomized comparison between continuing VKA or switching to aNOAC.

One trial proposed or ongoing with NOACs in patients with andwithout AF, is the Comparison of Antithrombotic Treatments AfterAortic Valve Replacement. Rivaroxaban: A New AntithromboticTreatment for Patients With Mechanical Prosthetic Aortic HeartValve: CATHAR trial(https://clinicaltrials.gov/ct2/show/NCT02128841?term=rivaroxabanþandþmechanicalþvalve&rank=2).

Bioprostheses, trans-aortic valveintervention, and transcatheter mitralvalve interventionsUsually, patients with a bioprosthesis and AF receive a VKA.Pericardial valves are less thrombogenic than mechanical valve pros-theses. Some physicians do not consider bioprostheses as a contra-indication of NOACs. Before recommending a NOAC rather thanVKA for these patients, a randomized trial is needed. This is also thecase for patients undergoing valve repair.

Trans-aortic valve intervention corresponds to transluminal implan-tation of a bioprosthesis and is being increasingly used. The antithrom-botic treatment in patients with sinus rhythm and TAVI remainscontroversial and the optimal treatment in patients with AF requiringTAVI (as well as TMVI—see earlier section) is currently unknown.

A global study comparing a rivaroxaban-based anti-thromboticstrategy to an antiplatelet-based strategy after TAVI to optimizeclinical outcomes (GALILEO) is currently ongoing.131 The twoarms consist of either rivaroxaban 10 mg once daily and aspirin75–100 mg for the first 90 days, followed by rivaroxaban alone; orclopidogrel 75 mg and aspirin 75–100 mg for the first 90 days, fol-lowed by clopidogrel alone. Patients with current or previous AFare excluded. The investigators assume that 15% of patients insinus rhythm at inclusion will develop AF during follow-up.Treatment after new onset AF will be, in patients randomized torivaroxaban, a switch to rivaroxaban 20 mg OD or 15 mg OD forthose with moderate renal impairment and in those randomizedto clopidogrel, a switch to VKA (target INR 2–3).

Another ongoing study is the Anti-Thrombotic Strategy afterTrans-Aortic Valve Implantation for Aortic Stenosis (ATLANTIS)study which is ongoing and plans to include 1509 patients aftersuccessful TAVI procedure. Randomization will be stratifiedaccording to the need for oral anticoagulant. Patients with an indi-cation for OAC will be randomized 1:1 to VKA or apixaban 5 mgb.i.d. The primary endpoint after 1 year follow-up is a compositeof death, myocardial infarction, stroke, systemic embolization,intracardiac or bioprosthesis thrombus, episode of deep veinthrombosis or pulmonary embolism, and major bleeding. Patientswith no indication for oral anticoagulant therapy will be random-ized 1:1 to either apixaban 5 mg bid or antiplatelet therapy. Othertrials are also proposed or ongoing with NOACs in patients withand without AF, include the RIvaroxaban for bioprostheticValvular Heart diseasE and atRial Fibrillation Trial (Warfarin vs.Rivaroxaban): RIVER Trial.

.................................................................................................

Coloured

heart

Supporting

references

• Appropriate use of oral anticoagu-

lants, when clinically indicated, has

both a clinical and economic value

since underutilization and/or non-

adherence are associated with

adverse outcomes and increased

costs.

121–123

Antithrombotic therapy in AF with valvular heart disease 1758o

Downloaded from https://academic.oup.com/europace/article-abstract/19/11/1757/4098134by gueston 25 August 2018

Page 18: Antithrombotic therapy inatrial fibrillation · 2019-04-12 · Antithrombotic therapy inatrial fibrillation associated withvalvular heart disease: a joint consensus document from

Native valve diseasesThe main phase III studies of NOACs have used variable criteria forexcluding valvular patients. Some studies (ROCKET-AF andARISTOTLE) only excluded patients with mechanical valve prosthe-ses and significant (moderate to severe) mitral stenosis. The sub-analyses did not show any differences in efficacy among patients withand without VHD. In the ROCKET-AF, there was more bleeding onrivaroxaban than on VKA in patients with VHD.

A report from the Loire Valley Atrial Fibrillation Project comparedthe outcome of patients without any valve disease and those withvalve disease but did not include either valve prosthesis or mitralstenosis. Although patients with VHD had a higher risk of stroke andTE events on univariable analysis, the difference was no longer signifi-cant after adjustment, in line with an older age and a higherCHA2DS2-VASC score in patients with VHD.128

However, post hoc analyses are only hypothesis generating. LargeRCTs are needed with NOACs in the setting of AS, non-rheumaticAR and MR before the role of NOACs can be fully defined in thissetting.

Mitral stenosisThere has not yet been a randomized trial comparing VKA andNOACs in these patients. The prevalence of rheumatic mitral steno-sis has become low in Western countries but remains high in EasternEurope, India, Africa, South America, and south East Asia. In theseregions, the time in therapeutic range is only 35–44%, according to aglobal AF registry.7 Randomized clinical trials comparing VKA with aNOAC is highly welcomed and should preferably include patientsfrom these affected countries.

Supplementary material

Supplementary material is available at Europace online.

References1. Rahman F, Kwan GF, Benjamin EJ. Global epidemiology of atrial fibrillation. Nat

Rev Cardiol 2014;11:639–54.2. Chugh SS, Havmoeller R, Narayanan K, Singh D, Rienstra M, Benjamin EJ et al.

Worldwide epidemiology of atrial fibrillation: a global burden of disease 2010study. Circulation 2014;129:837–47.

3. Molteni M, Polo Friz H, Primitz L, Marano G, Boracchi P, Cimminiello C. Thedefinition of valvular and non-valvular atrial fibrillation: results of a physicians’survey. Europace 2014;16:1720–5.

4. Kirchhof P, Benussi S, Kotecha D, Ahlsson A, Atar D, Casadei B et al. 2016 ESCGuidelines for the management of atrial fibrillation developed in collaborationwith EACTS: the Task Force for the management of atrial fibrillation of theEuropean Society of Cardiology (ESC) Developed with the special contributionof the European Heart Rhythm Association (EHRA) of the ESCEndorsed by theEuropean Stroke Organisation (ESO). Europace 2016;18:1609–78.

5. De Caterina R, John Camm A. Non-vitamin K antagonist oral anticoagulants inatrial fibrillation accompanying mitral stenosis: the concept for a trial. Europace2016;18:6–11.

6. De Caterina R, Camm AJ. What is 0valvular0 atrial fibrillation? A reappraisal. EurHeart J 2014;35:3328–35.

7. Oldgren J, Healey JS, Ezekowitz M, Commerford P, Avezum A, Pais P et al.Investigators R-LAFR. Variations in cause and management of atrial fibrillation ina prospective registry of 15,400 emergency department patients in 46 coun-tries: the RE-LY Atrial Fibrillation Registry. Circulation 2014;129:1568–76.

8. Fukuda N, Hirai T, Ohara K, Nakagawa K, Nozawa T, Inoue H. Relation of theseverity of mitral regurgitation to thromboembolic risk in patients with atrialfibrillation. Int J Cardiol 2011;146:197–201.

9. Heidbuchel H, Verhamme P, Alings M, Antz M, Diener HC, Hacke W et al.Advisors. Updated European Heart Rhythm Association Practical Guide on the

use of non-vitamin K antagonist anticoagulants in patients with non-valvularatrial fibrillation. Europace 2015;17:1467–507.

10. Petronio AS, Capranzano P, Barbato E, Piazza N, Baumbach A, Haude M et al.Current status of transcatheter mitral valve therapy in Europe: results from anEAPCI survey (Part II). EuroIntervention 2017;12:1934–9.

11. Elliott PM, Anastasakis A, Borger MA, Borggrefe M, Cecchi F, Charron P et al.2014 ESC Guidelines on diagnosis and management of hypertrophic cardiomy-opathy: the Task Force for the Diagnosis and Management of HypertrophicCardiomyopathy of the European Society of Cardiology (ESC). Eur Heart J2014;35:2733–79.

12. Benhorin J, Bodenheimer M, Brown M, Case R, Dwyer EM Jr, Eberly S et al.Improving clinical practice guidelines for practicing cardiologists. Am J Cardiol2015;115:1773–6.

13. Piccini JP, Hammill BG, Sinner MF, Jensen PN, Hernandez AF, Heckbert SRet al. Incidence and prevalence of atrial fibrillation and associated mortalityamong Medicare beneficiaries: 1993–2007. Circ Cardiovasc Qual Outcomes2012;5:85–93.

14. Friberg L, Bergfeldt L. Atrial fibrillation prevalence revisited. J Intern Med2013;274:461–8.

15. Krijthe BP, Kunst A, Benjamin EJ, Lip GY, Franco OH, Hofman A et al.Projections on the number of individuals with atrial fibrillation in the EuropeanUnion, from 2000 to 2060. Eur Heart J 2013;34:2746–51.

16. Lip GY, Brechin CM, Lane DA. The global burden of atrial fibrillation andstroke: a systematic review of the epidemiology of atrial fibrillation in regionsoutside North America and Europe. Chest 2012;142:1489–98.

17. De Caterina R, Husted S, Wallentin L, Andreotti F, Arnesen H, Bachmann Fet al. New oral anticoagulants in atrial fibrillation and acute coronary syn-dromes: ESC Working Group on Thrombosis-Task Force on Anticoagulants inHeart Disease position paper. J Am Coll Cardiol 2012;59:1413–25.

18. Watson T, Shantsila E, Lip GY. Mechanisms of thrombogenesis in atrial fibrilla-tion: Virchow’s triad revisited. Lancet 2009;373:155–66.

19. Goette A, Kalman JM, Aguinaga L, Akar J, Cabrera JA, Chen SA et al. EHRA/HRS/APHRS/SOLAECE expert consensus on atrial cardiomyopathies: definition,characterization, and clinical implication. Europace 2016;18:1455–90.

20. Baumgartner H, Hung J, Bermejo J, Chambers JB, Evangelista A, Griffin BP et al.;American Society of Echocardiography; European Association of Echocardiography.Echocardiographic assessment of valve stenosis: EAE/ASE recommendations for clin-ical practice. J Am Soc Echocardiogr 2009;22:1–23. quiz 101-2.

21. Fauchier L, Philippart R, Clementy N, Bourguignon T, Angoulvant D, Ivanes Fet al. How to define valvular atrial fibrillation? Arch Cardiovasc Dis2015;108:530–9.

22. Dewanjee MK, Gross DR, Zhai P, Lanzo S, Shim H, Park K et al.Thrombogenicity of polyethylene oxide-bonded Dacron sewing ring in a mech-anical heart valve. J Heart Valve Dis 1999;8:324–30.

23. Natorska J, Mazur P, Undas A. Increased bleeding risk in patients with aorticvalvular stenosis: from new mechanisms to new therapies. Thromb Res2016;139:85–9.

24. Alfieri O, Andreotti F, Antunes MJ, Bar�on-Esquivias G, Baumgartner H, BorgerMA et al. Guidelines on the management of valvular heart disease (version2012). Eur Heart J 2012;33:2451–96.

25. Torella M, Torella D, Chiodini P, Franciulli M, Romano G, De Santo L, De FeoM et al. LOWERing the INtensity of oral anticoaGulant Therapy in patientswith bileaflet mechanical aortic valve replacement: results from the“LOWERING-IT” Trial. Am Heart J 2010;160:171–8.

26. Carnicelli AP, De Caterina R, Halperin JL, Renda G, Ruff CT, Trevisan M et al.Edoxaban for the prevention of thromboembolism in patients with atrial fibrilla-tion and bioprosthetic valves. Circulation 2017;135:1273–5.

27. Renda R, De Caterina R, Carnicelli A, Nordio F, Mercuri M, Ruff C et al.Outcomes in 2824 patients with valvular heart disease treated with edoxa-ban or warfarin in the ENGAGE AF-TIMI 48 trial. J Am Coll Cardiol2016;67:2194.

28. De Caterina R, Renda G, Carnicelli AP, Nordio F, Trevisan M, Mercuri MF et al.Valvular heart disease patients on edoxaban or warfarin in the ENGAGE AF-TIMI 48 Trial. J Am Coll Cardiol 2017;69:1372–82.

29. Salem DN, O’gara PT, Madias C, Pauker SG. Valvular and structural heart dis-ease: American College of Chest Physicians evidence-based clinical practiceguidelines (8th edition). Chest 2008;133:593S–629S.

30. Heras M, Chesebro JH, Fuster V, Penny WJ, Grill DE, Bailey KR et al. High riskof thromboemboli early after bioprosthetic cardiac valve replacement. J Am CollCardiol 1995;25:1111–9.

31. Merie C, Køber L, Skov Olsen P, Andersson C, Gislason G, Skov Jensen J et al.Association of warfarin therapy duration after bioprosthetic aortic valve re-placement with risk of mortality, thromboembolic complications, and bleeding.JAMA 2012;308:2118–25.

32. January CT, Wann LS, Alpert JS, Calkins H, Cigarroa JE, Cleveland JC et al.;American College of Cardiology/American Heart Association Task Force on

1758p G.Y.H. Lip

Downloaded from https://academic.oup.com/europace/article-abstract/19/11/1757/4098134by gueston 25 August 2018

Page 19: Antithrombotic therapy inatrial fibrillation · 2019-04-12 · Antithrombotic therapy inatrial fibrillation associated withvalvular heart disease: a joint consensus document from

Practice Guidelines. 2014 AHA/ACC/HRS guideline for the management of pa-tients with atrial fibrillation: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines and the HeartRhythm Society. J Am Coll Cardiol 2014;64:e1–76.

33. Aramendi JI, Mestres C-A, Mestres C-A, Martinez-Le�on J, Campos V, Mu~noz Get al. Triflusal versus oral anticoagulation for primary prevention of thrombo-embolism after bioprosthetic valve replacement (trac): prospective, random-ized, co-operative trial. Eur J Cardio-Thorac Surg 2005;27:854–60.

34. Brueck M, Kramer W, Vogt P, Steinert N, Roth P, Gorlach G et al. Antiplatelettherapy early after bioprosthetic aortic valve replacement is unnecessary in pa-tients without thromboembolic risk factors. Eur J Cardiothorac Surg2007;32:108–12.

35. Philippart R, Brunet-Bernard A, Clementy N, Bourguignon T, Mirza A,Angoulvant D et al. Oral anticoagulation, stroke and thromboembolism in pa-tients with atrial fibrillation and valve bioprosthesis. The Loire Valley AtrialFibrillation Project. Thromb Haemost 2016;115:1056–63

36. Duraes AR, de Souza Roriz P, de Almeida Nunes B, Albuquerque FP, deBulhoes FV, de Souza Fernandes AM et al. Dabigatran versus warfarin after bio-prosthesis valve replacement for the management of atrial fibrillation postoper-atively: dawa pilot study. Drugs R D 2016;16:149–54.

37. Yadlapati A, Groh C, Malaisrie SC, Gajjar M, Kruse J, Meyers S et al. Efficacy andsafety of novel oral anticoagulants in patients with bioprosthetic valves. Clin ResCardiol 2016;105:268–72.

38. Ende G, Sichting L, Pfluecke C, Quick S, Schoener L, Strasser RH et al.Anticoagulation therapy of patients with atrial fibrillation after TAVI–DresdnerDOAK Register-TAVI (DDRT). Eur Heart J 2015;36:343.

39. Iung B, Baron G, Butchart EG, Delahaye F, Gohlke-B€arwolf C, Levang OW et al.A prospective survey of patients with valvular heart disease in Europe: the EuroHeart Survey on valvular heart disease. Eur Heart J 2003;24:1231–43.

40. Butchart EG, Gohlke-B€arwolf C, Antunes MJ, Tornos P, De Caterina R,Cormier B, et al.; Working Groups on Valvular Heart Disease Thrombosis,Cardiac Rehabilitation and Exercise Physiology, European Society ofCardiology. Recommendations for the management of patients after heart valvesurgery. Eur Heart J 2005;26:2463–71.

41. Camm AJ, Kirchhof P, Lip GYH, Schotten U, Savelieva I, Ernst S et al. Guidelinesfor the management of atrial fibrillation: the Task Force for the Management ofAtrial Fibrillation of the European Society of Cardiology (ESC). Europace2010;12:1360–420.

42. Nishimura RA, Otto CM, Bonow RO, Carabello BA, Erwin JP 3rd, Guyton RAet al. 2014 AHA/ACC Guideline for the Management of Patients With ValvularHeart Disease: executive summary: a report of the American College ofCardiology/American Heart Association Task Force on Practice Guidelines.Circulation 2014;129:2440–92.

43. Oxenham H, Bloomfield P, Wheatley DJ, Lee RJ, Cunningham J, Prescott RJet al. Twenty year comparison of a Bjork-Shiley mechanical heart valve withporcine bioprostheses. Heart 2003;89:715–21.

44. Whitlock RP, Sun JC, Fremes SE, Rubens FD, Teoh KH. American College of ChestP. Antithrombotic and thrombolytic therapy for valvular disease: antithrombotictherapy and prevention of thrombosis, 9th ed: American College of Chest PhysiciansEvidence-Based Clinical Practice Guidelines. Chest 2012;141:e576S–600S.

45. Turpie AG, Gent M, Laupacis A, Latour Y, Gunstensen J, Basile F et al. A com-parison of aspirin with placebo in patients treated with warfarin after heart-valve replacement. N Engl J Med 1993;329:524–9.

46. Laffort P, Roudaut R, Roques X, Lafitte S, Deville C, Bonnet J et al. Early andlong-term (one-year) effects of the association of aspirin and oral anticoagulanton thrombi and morbidity after replacement of the mitral valve with the St.Jude medical prosthesis: a clinical and transesophageal echocardiographic study.J Am Coll Cardiol 2000;35:739–46.

47. Camm AJ, Lip GY, De Caterina R, Savelieva I, Atar D, Hohnloser SH et al. 2012focused update of the ESC Guidelines for the management of atrial fibrillation:an update of the 2010 ESC Guidelines for the management of atrial fibrillation.Developed with the special contribution of the European Heart RhythmAssociation. Europace 2012;14:1385–413.

48. Massel DR, Little SH. Antiplatelet and anticoagulation for patients with pros-thetic heart valves. Cochrane Database Syst Rev 2013;7:Cd003464.

49. Larson RJ, Fisher ES. Should aspirin be continued in patients started on war-farin? J Gen Intern Med 2004;19:879–86.

50. Altman R, Rouvier J, Gurfinkel E, Scazziota A, Turpie AG. Comparison of high-dose with low-dose aspirin in patients with mechanical heart valve replacementtreated with oral anticoagulant. Circulation 1996;94:2113–6.

51. Meschengieser SS, Fondevila CG, Frontroth J, Santarelli MT, Lazzari MA. Low-intensity oral anticoagulation plus low-dose aspirin versus high-intensity oralanticoagulation alone: a randomized trial in patients with mechanical prostheticheart valves. J Thorac Cardiovasc Surg 1997;113:910–6.

52. Pengo V, Palareti G, Cucchini U, Molinatti M, Del Bono R, Baudo F et al. Low-intensity oral anticoagulant plus low-dose aspirin during the first six months

versus standard-intensity oral anticoagulant therapy after mechanical heart valvereplacement: a pilot study of low-intensity warfarin and aspirin in cardiac pros-theses (LIWACAP). Clin Appl Thromb Hemost 2007;13:241–8.

53. Dong MF, Ma ZS, Ma SJ, Chai SD, Tang PZ, Yao DK et al. Anticoagulation ther-apy with combined low dose aspirin and warfarin following mechanical heartvalve replacement. Thromb Res 2011;128:e91–4.

54. Dentali F, Douketis JD, Gianni M, Lim W, Crowther MA. Meta-analysis: anti-coagulant prophylaxis to prevent symptomatic venous thromboembolism inhospitalized medical patients. Ann Intern Med 2007;146:278–88.

55. Meschia JF, Bushnell C, Boden-Albala B, Braun LT, Bravata DM, Chaturvedi Set al. Guidelines for the primary prevention of stroke: a statement for health-care professionals from the American Heart Association/American StrokeAssociation. Stroke 2014;45:3754–832.

56. Nishimura RA, Otto CM, Bonow RO, Carabello BA, Erwin JP 3rd, Fleisher LAet al. 2017 AHA/ACC Focused Update of the 2014 AHA/ACC Guideline forthe Management of Patients With Valvular Heart Disease: a report of theAmerican College of Cardiology/American Heart Association Task Force onclinical practice guidelines. Circulation 2017;135:e1159–e1195.

57. Eikelboom JW, Hirsh J. Combined antiplatelet and anticoagulant therapy: clinicalbenefits and risks. J Thromb Haemost 2007;5:255–63.

58. Patel MR, Mahaffey KW, Garg J, Pan G, Singer DE, Hacke W, et al. Rivaroxabanversus warfarin in nonvalvular atrial fibrillation. N Engl J Med 2011;365:883–91.

59. Ezekowitz MD, Nagarakanti R, Noack H, Brueckmann M, Litherland C, JacobsM et al. Comparison of dabigatran and warfarin in patients with atrial fibrillationand valvular heart disease: the RE-LY Trial (randomized evaluation of long-termanticoagulant therapy). Circulation 2016;134:589–98.

60. Granger CB, Alexander JH, McMurray JJ, Lopes RD, Hylek EM, Hanna M et al.;Committees A, Investigators. Apixaban versus warfarin in patients with atrialfibrillation. N Engl J Med 2011;365:981–92.

61. Connolly SJ, Ezekowitz MD, Yusuf S, Eikelboom J, Oldgren J, Parekh A,Committee R-LS, Investigators et al. Dabigatran versus warfarin in patients withatrial fibrillation. N Engl J Med 2009;361:1139–51.

62. Giugliano RP, Ruff CT, Braunwald E, Murphy SA, Wiviott SD, Halperin JL et al.ENGAGE-AF TIMI48; Investigators. Edoxaban versus warfarin in patients withatrial fibrillation. N Engl J Med 2013;369:2093–104.

63. Connolly SJ, Eikelboom J, Joyner C, Diener HC, Hart R, Golitsyn S et al.Apixaban in patients with atrial fibrillation. N Engl J Med 2011;364:806–17.

64. Breithardt G, Baumgartner H, Berkowitz SD, Hellkamp AS, Piccini JP, StevensSR et al.; Committee RAS, Investigators. Clinical characteristics and outcomeswith rivaroxaban vs. warfarin in patients with non-valvular atrial fibrillation butunderlying native mitral and aortic valve disease participating in the ROCKETAF trial. Eur Heart J 2014;35:3377–85.

65. Avezum A, Lopes RD, Schulte PJ, Lanas F, Gersh BJ, Hanna M et al. Apixaban incomparison with warfarin in patients with atrial fibrillation and valvular heartdisease: findings from the Apixaban for Reduction in Stroke and OtherThromboembolic Events in Atrial Fibrillation (ARISTOTLE) Trial. Circulation2015;132:624–32.

66. Lip GY, Laroche C, Dan GA, Santini M, Kalarus Z, Rasmussen LH et al. A pro-spective survey in European Society of Cardiology member countries of atrialfibrillation management: baseline results of EURObservational ResearchProgramme Atrial Fibrillation (EORP-AF) pilot general registry. Europace2014;16:308–19.

67. Pokorney SDRM, Wojdyla DM, Gersh BJ, Lopes RD, Lewis BS, Hanna M et al.Apixaban use in patients with atrial fibrillation with bioprosthetic valves: insightsfrom ARISTOTLE. Circulation 2015;132:A17277.

68. Jaffer IH, Stafford AR, Fredenburgh JC, Whitlock RP, Chan NC, Weitz JI.Dabigatran is less effective than warfarin at attenuating mechanical heart valve-induced thrombin generation. J Am Heart Assoc 2015;4:e002322.

69. Breithardt G, Baumgartner H, Berkowitz SD, Hellkamp AS, Piccini JP,Lokhnygina Y, Committee RAS, Investigators et al. Native valve disease in pa-tients with non-valvular atrial fibrillation on warfarin or rivaroxaban. Heart2016;102:1036–43.

70. Renda G, Ricci F, Giugliano RP, De Caterina R. Non-vitamin K antagonist oralanticoagulants in patients with atrial fibrillation and valvular heart disease. J AmColl Cardiol 2017;69:1363–71.

71. Siontis KC, Yao X, Gersh BJ, Noseworthy PA. Direct oral anticoagulants in pa-tients with atrial fibrillation and valvular heart disease other than significant mi-tral stenosis and mechanical valves: a meta-analysis. Circulation 2017;135:714–6.

72. Thompson JL, Hamner CE, Potter DD, Lewin M, Sundt TM, Schaff HV.Melagatran for thromboprophylaxis after mechanical valve implantation: resultsin a heterotopic porcine model. J Thorac Cardiovasc Surg 2007;134:359–65.

73. McKellar SH, Abel S, Camp CL, Suri RM, Ereth MH, Schaff HV. Effectiveness ofdabigatran etexilate for thromboprophylaxis of mechanical heart valves. J ThoracCardiovasc Surg 2011;141:1410–6.

74. Schomburg JL, Medina EM, Lahti MT, Bianco RW. Dabigatran versus warfarin aftermechanical mitral valve replacement in the swine model. J Invest Surg 2012;25:150–5.

Antithrombotic therapy in AF with valvular heart disease 1758q

Downloaded from https://academic.oup.com/europace/article-abstract/19/11/1757/4098134by gueston 25 August 2018

Page 20: Antithrombotic therapy inatrial fibrillation · 2019-04-12 · Antithrombotic therapy inatrial fibrillation associated withvalvular heart disease: a joint consensus document from

75. Eikelboom JW, Connolly SJ, Brueckmann M, Granger CB, Kappetein AP, MackMJ et al.; Investigators R-A. Dabigatran versus warfarin in patients with mechan-ical heart valves. N Engl J Med 2013;369:1206–14.

76. Connolly SJ, Pogue J, Eikelboom J, Flaker G, Commerford P, Franzosi MG et al.;AW Investigators. Benefit of oral anticoagulant over antiplatelet therapy in atrialfibrillation depends on the quality of international normalized ratio controlachieved by centers and countries as measured by time in therapeutic range.Circulation 2008;118:2029–37.

77. Nombela-Franco L, Webb JG, de Jaegere PP, Toggweiler S, Nuis RJ, Dager AEet al. Timing, predictive factors, and prognostic value of cerebrovascular eventsin a large cohort of patients undergoing transcatheter aortic valve implantation.Circulation 2012;126:3041–53.

78. Stortecky S, Buellesfeld L, Wenaweser P, Heg D, Pilgrim T, Khattab AA et al. Atrialfibrillation and aortic stenosis: impact on clinical outcomes among patients undergo-ing transcatheter aortic valve implantation. Circ Cardiovasc Interv 2013;6:77–84.

79. Holmes DR Jr, Mack MJ, Kaul S, Agnihotri A, Alexander KP, Bailey SR et al.;American Heart A, American Society of E, European Association for Cardio-Thoracic S, Heart Failure Society of A, Mended H, Society of Cardiovascular A,Society of Cardiovascular Computed T, Society for Cardiovascular MagneticResearcher. 2012 ACCF/AATS/SCAI/STS expert consensus document ontranscatheter aortic valve replacement: developed in collaboration with theAmerican Heart Association, American Society of Echocardiography, EuropeanAssociation for Cardio-Thoracic Surgery, Heart Failure Society of America,Mended Hearts, Society of Cardiovascular Anesthesiologists, Society ofCardiovascular Computed Tomography, and Society for CardiovascularMagnetic Resonance. J Thorac Cardiovasc Surg 2012;144:e29–84.

80. Lip GY, Windecker S, Huber K, Kirchhof P, Marin F, Ten Berg JM et al.Management of antithrombotic therapy in atrial fibrillation patients presentingwith acute coronary syndrome and/or undergoing percutaneous coronary orvalve interventions: a joint consensus document of the European Society ofCardiology Working Group on Thrombosis, European Heart RhythmAssociation (EHRA), European Association of Percutaneous CardiovascularInterventions (EAPCI) and European Association of Acute Cardiac Care(ACCA) endorsed by the Heart Rhythm Society (HRS) and Asia-Pacific HeartRhythm Society (APHRS). Eur Heart J 2014;35:3155–79.

81. Webb J, Rodes-Cabau J, Fremes S, Pibarot P, Ruel M, Ibrahim R et al.Transcatheter aortic valve implantation: a Canadian Cardiovascular Society pos-ition statement. Can J Cardiol 2012;28:520–8.

82. Cayla G, Hulot JS, O’connor SA, Pathak A, Scott SA, Gruel Y et al. Clinical,angiographic, and genetic factors associated with early coronary stent throm-bosis. JAMA 2011;306:1765–74.

83. Abdul-Jawad Altisent O, Durand E, Munoz-Garcia AJ, Nombela-Franco L,Cheema A, Kefer J et al. Warfarin and antiplatelet therapy versus warfarin alonefor treating patients with atrial fibrillation undergoing transcatheter aortic valvereplacement. JACC Cardiovasc Interv 2016;9:1706–17.

84. Dewilde WJ, Oirbans T, Verheugt FW, Kelder JC, De Smet BJ, Herrman JP et al.;WOEST study investigators. Use of clopidogrel with or without aspirin in patientstaking oral anticoagulant therapy and undergoing percutaneous coronary interven-tion: an open-label, randomised, controlled trial. Lancet 2013;381:1107–15.

85. Schulz-Schupke S, Byrne RA, Ten Berg JM, Neumann FJ, Han Y, Adriaenssens Tet al. EFoMDATAD-EST Investigators. ISAR-SAFE: a randomized, double-blind,placebo-controlled trial of 6 vs. 12 months of clopidogrel therapy after drug-eluting stenting. Eur Heart J 2015;36:1252–63.

86. Urban P, Meredith IT, Abizaid A, Pocock SJ, Carrie D, Naber C et al. Polymer-free drug-coated coronary stents in patients at high bleeding risk. N Engl J Med2015;373:2038–47.

87. Gargiulo G, Collet JP, Valgimigli M. Antithrombotic therapy in TAVI patients:changing concepts. EuroIntervention 2015;11:W92–5.

88. Makkar RR, Fontana G, Jilaihawi H, Chakravarty T, Kofoed KF, de Backer Oet al. Possible subclinical leaflet thrombosis in bioprosthetic aortic valves. N EnglJ Med 2015;373:2015–24.

89. Seeger J, Gonska B, Rodewald C, Rottbauer W, Wohrle J. Apixaban in patientswith atrial fibrillation after transfemoral aortic valve replacement. JACCCardiovasc Interv 2017;10:66–74.

90. Holmes DR Jr, Kar S, Price MJ, Whisenant B, Sievert H, Doshi SK et al.Prospective randomized evaluation of the watchman left atrial appendage clos-ure device in patients with atrial fibrillation versus long-term warfarin therapy:the PREVAIL trial. J Am Coll Cardiol 2014;64:1–12.

91. Holmes DR, Reddy VY, Turi ZG, Doshi SK, Sievert H, Buchbinder M, PAInvestigators et al. Percutaneous closure of the left atrial appendage versus war-farin therapy for prevention of stroke in patients with atrial fibrillation: a rando-mised non-inferiority trial. Lancet 2009;374:534–42.

92. Price MJ, Reddy VY, Valderrabano M, Halperin JL, Gibson DN, Gordon N et al.Bleeding outcomes after left atrial appendage closure compared with long-termwarfarin: a pooled, patient-level analysis of the WATCHMAN RandomizedTrial Experience. JACC Cardiovasc Interv 2015;8:1925–32.

93. Meier B, Blaauw Y, Khattab AA, Lewalter T, Sievert H, Tondo C et al. EHRA/EAPCI expert consensus statement on catheter-based left atrial appendageocclusion. Europace 2014;16:1397–416.

94. Bosche LI, Afshari F, Schone D, Ewers A, Mugge A, Gotzmann M. Initial experi-ence with novel oral anticoagulants during the first 45 days after left atrial ap-pendage closure with the watchman device. Clin Cardiol 2015;38:720–4.

95. Reddy VY, Mobius-Winkler S, Miller MA, Neuzil P, Schuler G, Wiebe J et al.Left atrial appendage closure with the Watchman device in patients with acontraindication for oral anticoagulation: the ASAP study (ASA Plavix FeasibilityStudy With Watchman Left Atrial Appendage Closure Technology). J Am CollCardiol 2013;61:2551–6.

96. Bax JJ, Delgado V, Bapat V, Baumgartner H, Collet JP, Erbel R et al. Open issues intranscatheter aortic valve implantation. Part 2: Procedural issues and outcomesafter transcatheter aortic valve implantation. Eur Heart J 2014;35:2639–54.

97. Sliwa K, Johnson MR, Zilla P, Roos-Hesselink JW. Management of valvular dis-ease in pregnancy: a global perspective. Eur Heart J 2015;36:1078–89.

98. van Hagen IM, Roos-Hesselink JW, Ruys TP, Merz WM, Goland S, Gabriel Het al. Pregnancy in women with a mechanical heart valve: data of the EuropeanSociety of Cardiology Registry of Pregnancy and Cardiac Disease (ROPAC).Circulation 2015;132:132–42.

99. Xu Z, Fan J, Luo X, Zhang WB, Ma J, Lin YB et al. Anticoagulation regimens dur-ing pregnancy in patients with mechanical heart valves: a systematic review andmeta-analysis. Can J Cardiol 2016;32:1248 e1–e9.

100. Vahanian A, Alfieri O, Andreotti F, Antunes MJ, Baron-Esquivias G, BaumgartnerH et al. Guidelines ESCCfP, Joint Task Force on the Management of ValvularHeart Disease of the European Society of C, European Association for Cardio-Thoracic S. Guidelines on the management of valvular heart disease (version2012): the Joint Task Force on the Management of Valvular Heart Disease of theEuropean Society of Cardiology (ESC) and the European Association for Cardio-Thoracic Surgery (EACTS). Eur J Cardiothorac Surg 2012;42:S1–44.

101. Desai CS, Bonow RO. Transcatheter valve replacement for aortic stenosis: bal-ancing benefits, risks, and expectations. JAMA 2012;308:573–4.

102. Tillquist MN, Maddox TM. Cardiac crossroads: deciding between mechanical orbioprosthetic heart valve replacement. Patient Prefer Adherence 2011;5:91–9.

103. Lindman BR, Alexander KP, O’gara PT, Afilalo J. Futility, benefit, and transcath-eter aortic valve replacement. JACC Cardiovasc Interv 2014;7:707–16.

104. Charles C, Gafni A, Whelan T. Shared decision-making in the medical encounter:what does it mean? (or it takes at least two to tango). Soc Sci Med 1997;44:681–92.

105. Charles C, Gafni A, Whelan T. How to improve communication between doc-tors and patients. Learning more about the decision making context is import-ant. BMJ 2000;320:1220–1.

106. Malloy-Weir LJ, Charles C, Gafni A, Entwistle VA. Empirical relationships be-tween health literacy and treatment decision making: a scoping review of the lit-erature. Patient Educ Couns 2015;98:296–309.

107. Coylewright M, Palmer R, O’neill ES, Robb JF, Fried TR. Patient-defined goalsfor the treatment of severe aortic stenosis: a qualitative analysis. Health Expect2015.

108. Hussain AI, Garratt AM, Brunborg C, Aakhus S, Gullestad L, Pettersen KI.Eliciting patient risk willingness in clinical consultations as a means of improvingdecision-making of aortic valve replacement. J Am Heart Assoc 2016;4:e002828.

109. Birkmeyer NJ, Birkmeyer JD, Tosteson AN, Grunkemeier GL, Marrin CA,O’Connor GT. Prosthetic valve type for patients undergoing aortic valve re-placement: a decision analysis. Ann Thorac Surg 2000;70:1946–52.

110. Takkenberg JJ, Puvimanasinghe JP, van Herwerden LA, Eijkemans MJ, SteyerbergEW, Habbema JD et al. Decision-making in aortic valve replacement: bileafletmechanical valves versus stented bioprostheses. Neth Heart J 2003;11:5–10.

111. Narasimhan CVJ, Kishore AGR, Singh B, Dani S, Chawala K et al. The REALIZEAF study. An International, observational cross sectional survey evaluating AFManagement and Cardiovascular Risk profile of AF patients, Indian subset datapresented at the ISE Meeting Jaipur. 2012.

112. Zuhlke L, Engel ME, Karthikeyan G, Rangarajan S, Mackie P, Cupido B et al.Characteristics, complications, and gaps in evidence-based interventions inrheumatic heart disease: the Global Rheumatic Heart Disease Registry (theREMEDY study). Eur Heart J 2015;36:1115–22a.

113. Narasimhan C, Verma JS, Ravi Kishore AG, Singh B, Dani S, Chawala K et al.Cardiovascular risk profile and management of atrial fibrillation in India: realworld data from RealiseAF survey. Indian Heart J 2016;68:663–70.

114. Egbe AC, Pislaru SV, Pellikka PA, Poterucha JT, Schaff HV, Maleszewski JJ,Connolly HM. Bioprosthetic valve thrombosis versus structural failure: clinicaland echocardiographic predictors. 2015;66:2285–94.

115. Vora A, Kapoor A, Nair M, Lokhandwala Y, Narsimhan C, Ravikishore AG et al.Clinical presentation, management, and outcomes in the Indian Heart RhythmSociety-Atrial Fibrillation (IHRS-AF) registry. Indian Heart J 2017;69:43–7.

116. Marcolino MS, Polanczyk CA, Bovendorp AC, Marques NS, Silva LA, Turquia CP et al.Economic evaluation of the new oral anticoagulants for the prevention of thrombo-embolic events: a cost-minimization analysis. Sao Paulo Med J 2016;134:322–9.

1758r G.Y.H. Lip

Downloaded from https://academic.oup.com/europace/article-abstract/19/11/1757/4098134by gueston 25 August 2018

Page 21: Antithrombotic therapy inatrial fibrillation · 2019-04-12 · Antithrombotic therapy inatrial fibrillation associated withvalvular heart disease: a joint consensus document from

117. Maniadakis N, Vardas P, Mantovani LG, Fattore G, Boriani G. Economic evalu-ation in cardiology. Europace 2011;13:ii3–8.

118. Boriani G, Maniadakis N, Auricchio A, Muller-Riemenschneider F, Fattore G,Leyva F et al. Health technology assessment in interventional electrophysiologyand device therapy: a position paper of the European Heart RhythmAssociation. Eur Heart J 2013;34:1869–74.

119. Wolowacz SE, Samuel M, Brennan VK, Jasso-Mosqueda JG, Van Gelder IC. Thecost of illness of atrial fibrillation: a systematic review of the recent literature.Europace 2011;13:1375–85.

120. Boriani G, Diemberger I, Biffi M, Martignani C. Balancing the risk of hemorrhagevs thromboembolism in patients with atrial fibrillation: how to navigate betweenScylla and Charybdis? Chest 2010;138:1032–3.

121. Kirchhof P, Breithardt G, Bax J, Benninger G, Blomstrom-Lundqvist C, BorianiG et al. A roadmap to improve the quality of atrial fibrillation management: pro-ceedings from the fifth Atrial Fibrillation Network/European Heart RhythmAssociation consensus conference. Europace 2016;18:37–50.

122. Bruggenjurgen B, Rossnagel K, Roll S, Andersson FL, Selim D, Muller-NordhornJ et al. The impact of atrial fibrillation on the cost of stroke: the berlin acutestroke study. Value Health 2007;10:137–43.

123. Cotte FE, Chaize G, Kachaner I, Gaudin AF, Vainchtock A, Durand-Zaleski I.Incidence and cost of stroke and hemorrhage in patients diagnosed with atrialfibrillation in France. J Stroke Cerebrovasc Dis 2014;23:e73–83.

124. Mercaldi CJ, Siu K, Sander SD, Walker DR, Wu Y, Li Q et al. Long-term costs of ischemic stroke and major bleeding events among

medicare patients with nonvalvular atrial fibrillation. Cardiol Res Pract2012;2012:645469.

125. Casciano JP, Dotiwala ZJ, Martin BC, Kwong WJ. The costs of warfarin under-use and nonadherence in patients with atrial fibrillation: a commercial insurerperspective. JMCP 2013;19:302–16.

126. Beyer-Westendorf J, Ehlken B, Evers T. Real-world persistence and adherenceto oral anticoagulation for stroke risk reduction in patients with atrial fibrilla-tion. Europace 2016;18:1150–7.

127. Boriani G, Cimaglia P, Fantecchi E, Mantovani V, Ziacchi M, Valzania C et al. Non-valvular atrial fibrillation: potential clinical implications of the heterogeneous defin-itions used in trials on new oral anticoagulants. J Cardiovasc Med 2015;16:491–6.

128. Philippart R, Brunet-Bernard A, Clementy N, Bourguignon T, Mirza A, BabutyD et al. Prognostic value of CHA2DS2-VASc score in patients with 0non-valvu-lar atrial fibrillation0 and valvular heart disease: the Loire Valley AtrialFibrillation Project. Eur Heart J 2015;36:1822–30.

129. Kasmeridis C, Apostolakis S, Ehlers L, Rasmussen LH, Boriani G, Lip GY. Costeffectiveness of treatments for stroke prevention in atrial fibrillation: focus onthe novel oral anticoagulants. Pharmacoeconomics 2013;31:971–80.

130. Janzic A, Kos M. Cost effectiveness of novel oral anticoagulants for stroke pre-vention in atrial fibrillation depending on the quality of warfarin anticoagulationcontrol. Pharmacoeconomics 2015;33:395–408.

131. Hemmrich M, Peterson ED, Thomitzek K, Weitz JI. Spotlight on unmet needs instroke prevention: the PIONEER AF-PCI, NAVIGATE ESUS and GALILEOtrials. Thromb Haemost 2016;116:S33–40.

Antithrombotic therapy in AF with valvular heart disease 1758s

Downloaded from https://academic.oup.com/europace/article-abstract/19/11/1757/4098134by gueston 25 August 2018


Recommended